Next Article in Journal
Bioprospecting of Sea Anemones (Cnidaria, Anthozoa, Actiniaria) for β-Defensin-like α-Amylase Inhibitors
Next Article in Special Issue
A Rare Case of Anca Positivity and Antiphospholipid Antibodies in a Patient with Takayasu Arteritis: Case Report and Review of the Literature
Previous Article in Journal
Ion Channel Genes in Painful Neuropathies
Previous Article in Special Issue
Thrombosis and Hyperinflammation in COVID-19 Acute Phase Are Related to Anti-Phosphatidylserine and Anti-Phosphatidylinositol Antibody Positivity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Trophoblast Cell Function in the Antiphospholipid Syndrome

by
Svetlana Vrzić Petronijević
1,*,
Aleksandra Vilotić
2,
Žanka Bojić-Trbojević
2,
Sanja Kostić
1,
Miloš Petronijević
1,
Ljiljana Vićovac
2 and
Milica Jovanović Krivokuća
2,*
1
University of Belgrade, Faculty of Medicine, University Clinical Center of Serbia Clinic for Obstetrics and Gynecology, Koste Todorovića 26, 11000 Belgrade, Serbia
2
University of Belgrade, Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, Banatska 31b, 11080 Belgrade, Serbia
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(10), 2681; https://doi.org/10.3390/biomedicines11102681
Submission received: 30 July 2023 / Revised: 26 September 2023 / Accepted: 27 September 2023 / Published: 30 September 2023
(This article belongs to the Special Issue Basic and Clinical Researches of Antiphospholipid Syndrome)

Abstract

:
Antiphospholipid syndrome (APS) is a complex thrombo-inflammatory autoimmune disease characterized by the presence of antiphospholipid antibodies (aPL). Women with APS are at high risk of recurrent early pregnancy loss as well as late obstetrical complications—premature birth due to placental insufficiency or severe preeclampsia. Accumulating evidence implies that vascular thrombosis is not the only pathogenic mechanism in obstetric APS, and that the direct negative effect of aPL on the placental cells, trophoblast, plays a major role. In this review, we summarize the current findings regarding the potential mechanisms involved in aPL-induced trophoblast dysfunction. Introduction on the APS and aPL is followed by an overview of the effects of aPL on trophoblast—survival, cell function and aPL internalization. Finally, the implication of several non-coding RNAs in pathogenesis of obstetric APS is discussed, with special emphasis of their possible role in trophoblast dysfunction and the associated mechanisms.

1. Introduction

Antiphospholipid syndrome (APS) is a systemic autoimmune disorder characterized by recurrent arterial, venous and microvasculature thrombosis and/or obstetrical complications associated with circulating antiphospholipid antibodies (aPL) [1,2]. The diagnosis and classification of APS is based on the Sydney 2006 updated international classification criteria consensus [3]. According to these criteria, at least one of the clinical conditions and persistent detection of at least one of the criteria aPL have to be present for APS diagnosis [3] (Table 1). The autoantibodies accepted for the laboratory criteria include lupus anticoagulant, anticardiolipin and anti-β2-glycoprotein I IgG and/or IgM antibodies (Table 1). APS can be an isolated disease when it is defined as primary. Secondary APS represents coexistence of APS with some other autoimmune disorder, usually systemic lupus erythematosus (SLE) [1,4]. APS/SLE patients account for around 30% of all APS cases [5,6,7,8].
The estimated APS annual incidence and prevalence in the general population ranges between 1 and 2 cases per 100,000 persons and between 40 and 50 per 100,000 persons, respectively [9]. Most of the APS patients are diagnosed during the reproductive period with the mean age of diagnosis between 30 and 40 years for women, as several studies presented [6,8,9,10,11]. Moreover, APS is found to be more frequent in females especially when considering patients with secondary APS associated with SLE [6,9,10]. However, some studies found that there was no difference in APS frequency between sexes [7,8,9].
According to the clinical manifestations, two main subtypes of APS could be distinguished: vascular and obstetrical APS [12]. Vascular APS is mainly characterized by venous, arterial and small vessel thrombotic events in different organs [12]. Obstetrical APS (OAPS) is manifested with pregnancy morbidities and lower frequency of thrombotic events [12,13,14,15]. Distinct molecular signatures in these two APS subtypes were also found [16]. The most prevalent pregnancy complications in OAPS are early recurrent pregnancy loss (RPL), unexplained fetal death and stillbirth [5,14,17,18]. Complications in later stages of pregnancy including premature birth, preeclampsia (PE) and intrauterine growth restriction (IUGR) are also common for OAPS patients [5,14,17,18]. The original historic assumption was that complications in OAPS were associated with placental thrombotic phenomena [19]. However, experimental data accumulating over the past couple of decades have shown that inadequate placentation due to multiple detrimental effects of aPL on trophoblast, specialized placental cells, as well as other cell types of the placenta and uterus is a major cause of pregnancy morbidities in OAPS [12,14].
The gold standard treatment of APS is low dose aspirin combined with low molecular weight heparin at prophylactic or therapeutic doses, depending on a history of blood clots and previous complications during pregnancy [20,21]. In about 20–30% of OAPS patients, standard treatment does not give satisfactory results and they suffer from recurrent pregnancy complications [22]. There are several treatment options reserved for refractory OAPS including hydroxychloroquine, low-prednisone dose, intravenous immunoglobulins or plasma exchange [21]. Biologic therapies using anti-TNF-α antibodies in combination with standard treatment gave promising results for the treatment of refractory OAPS [22,23]. Recently, aPL-induced epigenetic modifications, including dysregulated expression of non-coding RNAs, emerged as key contributors to the APS progression as well as potential additional biomarkers and therapeutic targets in APS [24,25].
In this review, following the Introduction, we will briefly present general information on aPL types, their antigens and general mechanisms of action. Further, we will focus on aPL-induced effects on trophoblast cell survival and function. Finally, we will present current knowledge on non-coding RNAs as mediators of aPL-induced obstetric complications.

2. Antiphospholipid Antibodies

Antiphospholipid antibodies (aPL) are autoantibodies directed against phospholipids and/or phospholipid binding proteins present on cell membranes of various cell types, such as endothelial cells, leukocytes and platelets [26]. Laboratory criteria aPL, as mentioned above, include lupus anticoagulant, anticardiolipin and anti-β2-glycoprotein I (anti-β2GPI) antibodies [3]. In addition to these autoantibodies, aPL also include antibodies to annexin A5 [27], annexin A2 [28], protein S [29,30], phosphatidylethanolamine [31], lysobisphosphatidic acid [32], prothrombin [33] as well as autoantibodies to complexes, such as prothrombin/phosphatidylserine [34] and vimentin/cardiolipin [35], designated as non-criteria aPL. It was estimated that women with poor obstetric outcomes in 6-30% of all cases are carriers of aPL, either criteria and/or non-criteria [36,37,38,39].
A recent APS ACTION study has shown that a clinically meaningful aPL profile with positivity of all three laboratory criteria is associated with pronounced clinical features and more durable aPL for a period of 5 years in 78% of patients [40]. Within a spectrum of patients with clinical manifestations of APS, a significant portion was found not to meet these laboratory criteria. Patients without detectable criteria aPL are referred to as seronegative [41], while others that did not fulfill the aPL laboratory criteria regarding level or type of immunoglobulins are recognized as patients with non-criteria aPL, and/or lower level of criteria aPL [42].
Recently, regarding clinical obstetric manifestations, not much difference was noted in the cumulative incidence of adverse obstetrical events in seronegative and seropositive APS patients, although higher rates of intrauterine deaths (15% vs. 5%; p = 0.03), of PE (7% vs. 16%, p = 0.048) and lower live birth term (36 ± 3 vs. 38 ± 3 weeks of gestation; p = 0.04) were noted in seropositive APS patients [43]. The cumulative incidence of pregnancy complications was significantly decreased in treated versus untreated women with seronegative APS. A systematic review of studies comparing APS patients fulfilling Sydney criteria (definite APS) and non-criteria APS patients (NC-APS) presented that most studies have shown no significant difference in prevalence of clinical manifestations between definite and NC-APS patients including pregnancy morbidities [44]. A recent review focused on the evidence showing that non-criteria aPL may play a functional role in the signal transduction pathway(s) leading to thrombosis and pregnancy morbidity in seronegative APS patients [26]. The results of the recent retrospective multicenter study [45] from the European Registry on Obstetric Antiphospholipid Syndrome aimed at comparing clinical features, laboratory data and fetal–maternal outcomes between women with OAPS and with aPL-related obstetric complications not fulfilling Sydney criteria, yielded interesting conclusions pertinent to pregnancy outcomes in affected women that received treatment irrespective of fulfillment of aPL criteria. Treatment resulted in favorable obstetric outcomes for both the mother and the infant with no difference between OAPS and NC-OAPS. Given the shown obstetric benefits of treatment of patients with any aPL with clinical symptoms [43], it is worth stressing that OAPS, as the most frequent treatable autoimmune disease during pregnancy, should be treated irrespective of the fulfillment of the laboratory criteria.
Although accumulated data have shown a strong association of aPL and thrombosis, the underlying molecular mechanisms have not been completely elucidated and multiple mechanisms may be involved. It has been shown that aPL activates endothelial cells and promotes a proinflammatory and procoagulant cell phenotype through activation of Toll-like receptor 4/myeloid differentiation primary response 88 (TLR4/MyD88) signaling pathway, leading to the increased expression of adhesion molecules and release of cytokines [26,46]. Another mechanism of endothelial and monocyte cell activation by anti-β2GPI involves the upregulation of Tissue Factor which is a key molecule in extrinsic coagulation cascade initiation [47]. Platelets may also be activated by aPL, which leads to increased thromboxane A2 synthesis, glycoprotein IIb-IIIa expression and platelet factor-4 secretion [26]. It has also been demonstrated that annexin A5 protein functions as a physiological anticoagulant. It binds to phosphatidylserine on the cell surface forming a shield and prevents activation of procoagulant complexes [48].
In placental tissue, trophoblast cells abundantly express β2GPI [49], which together with hormonal and vascular changes linked to pregnancy highly contribute to the development of obstetrical complications in the presence of aPL. Besides trophoblast, decidual cells and other cell types at the feto–maternal interface highly express aPL antigens and can be affected by aPL as well. However, several lines of evidence described different roles of aPL in early and late pregnancy. Thus, in early pregnancy, aPL affect placentation and apoptosis of trophoblast cells, while thrombotic mechanisms are connected to late obstetric complications, such as IUGR and PE [1,50]. Moreover, it has been shown that anti-β2GPI binding to endothelial and trophoblast cells destroy the anticoagulant shield of annexin A5, thereby inducing a procoagulant state in placenta which can lead to thrombosis, thus influencing embryo fate [46,51].
At the level of trophoblast cells and the feto–maternal interface, various molecular mechanisms of aPL’s detrimental effects were proposed. Through the activation of TLRs and the NLR family pyrin domain containing 3 (NLRP3) inflammasome, aPL increase IL-1β and IL-8 trophoblast secretion [52,53]. It has been reported that aPL reduce beta-human chorionic gonadotropin (βhCG) production [54,55], signal transducer and activator of transcription 3 (STAT3) activity and interleukin (IL)-6 secretion, leading to decreased trophoblast function [56]. Several studies demonstrated a complement activation by aPL, which leads to the release of reactive oxygen species, antiangiogenic factors, Tissue Factor as well as TNF-α [57,58,59]. Due to the action of aPL, placentas of APS patients are structurally modified as a consequence of aPL internalization by syncytiotrophoblast [60], which will be discussed in the next section.
Clearly, there are many different mechanisms involved in APS during pregnancy. Therefore, the knowledge about different molecular mechanisms triggered by aPL involved in the abnormal development of placenta and placental dysfunction was and will be mostly useful in terms of OAPS management.

3. The Impact of aPL on Trophoblast Cells

Placenta is a unique organ essential for pregnancy success, which forms contact between the mother and fetus and exerts multiple important functions. Aberrant placentation is associated with diverse pregnancy complications such as miscarriage, stillbirth, pre-term labor, IUGR and PE [61]. Trophoblast cells are specific placental cells, exerting a variety of functions at the feto–maternal interface. Placental chorionic villi are covered with syncytiotrophoblast, a multinucleated trophoblast layer directly facing maternal circulation, which facilitates nutrient transport and gas exchange between mother and fetus. Syncytiotrophoblast secretes hormones necessary for the maintenance of healthy pregnancy such as βhCG and placental lactogen [61]. Underneath syncytium, there is a layer of cytotrophoblast cells which continuously proliferate and through constant fusion form syncytiotrophoblast. Another type of trophoblast cells are extravillous trophoblast cells (EVTs), which also differentiate from cytotrophoblast through the process of epithelial-mesenchymal transition (EMT). Detaching from the tips of the anchoring villi, EVTs invade the maternal decidual stroma, spiral arteries and other luminal structures in the uterus, which is essential for the process of placentation [61,62] (Figure 1A). The outcome of this process is adequately attached placenta and modified spiral arteries that enable sufficient supply of oxygen, nutrients and other factors to the developing fetus.
Trophoblast cells abundantly express aPL antigens, especially β2GPI [49]. Anti-β2GPI antibodies are considered to be the main contributors to the pathogenesis of OAPS and a great number of in vitro studies deciphering pathogenic mechanisms of the syndrome were conducted using these autoantibodies. It was found that aPL induce detrimental effects on trophoblast cells affecting their proliferation, differentiation and survival [63,64], as well as other cellular processes including invasion and migration [65,66,67,68,69] through different molecular mechanisms which will be discussed in this section.

3.1. Trophoblast Survival

Apoptosis has an important role in normal placental development [70,71]. However, increased rates of trophoblast apoptosis are associated with placental diseases [70,71]. In the past two decades, a growing body of evidence suggests that aPL affect trophoblast cell proliferation and apoptosis. In vitro studies on rat’s embryos demonstrated that purified aPL IgG treatment inhibited embryo and yolk sac growth and increased apoptosis of ectoplacental cone giant cells [72,73]. In mice, passive immunization with human aPL during gestation attenuated placental morphogenesis [69,74], reduced trophoblast proliferation [74] and increased placental apoptosis as evidenced by the increased index in TUNEL-positive cells and pronounced DNA-fragmentation [75].
Analyzing sections of human first trimester placentas after elective abortions and aPL-associated miscarriages, Bose et al. found that aPL-associated trophoblast development is aberrant [76]. Authors hypothesized that aPL may stimulate the premature onset of cytotrophoblast proliferation and differentiation, in favor of syncytial fusion, which could result in accelerated exhaustion of cytotrophoblast ‘stem’ cells, leading to the altered morphology and function of placenta and consequential pregnancy loss [76]. Furthermore, in vitro experiments using human placental explants from the first trimester of pregnancy cultured with sera of APS/SLE patients with history of RPL showed reduced placental villi growth and trophoblast cell proliferation as well as increased trophoblast apoptosis in treated explants [73,77]. During pregnancy, as part of normal placental aging, the syncytiotrophoblast layer sheds through the process of apoptosis releasing multinucleated syncytial aggregates known as trophoblast debris in maternal circulation [78]. Studies on the human placental explant model showed that aPL treatment increased trophoblast shedding rates and altered the cell death process through which trophoblast debris is formed [60,79]. It has been shown that aPL antibodies affected mitochondrial function through mitochondrial leakage and cytochrome C release, which eventually led to necrotic cell death and extrusion of necrotic trophoblast debris [60]. After phagocytosis, aPL-induced trophoblast debris stimulated activation of endothelial cells which could explain, at least in part, how aPL could increase the risk of PE and other adverse pregnancy outcomes associated with an activated endothelium in OAPS patients [60,79]. An additional proposed mechanism by which aPL could cause cell death in the syncytiotrophoblast is the interruption of placental lipid signaling and decreased expression of protein kinase C-epsilon (PRKCE) as determined on the placental explant model [80].
Moreover, aPL-induced transcriptome and metabolome alterations connected to trophoblast cell death have been found [80,81]. Transcriptomic analysis revealed changes in the expression of factors involved in the regulation of apoptosis, including BCL2L1, MCL1, PDCD2L, FASLG, SEMA6A, PRKCE and TRAIL mRNAs in response to aPL treatment of human placental explants [81]. Altered lipid metabolism, especially of ceramides and diacylglycerols, important players in cell death regulatory pathways, was the most pronounced aPL-induced metabolic change detected in treated human placental explants [80]. Expression of anti-apoptotic BCL2 and pro-apoptotic BAX genes on mRNA and protein levels was altered in primary trophoblast cells after aPL treatment [82]. Reduced BCL2/BAX ratio indicating a pro-apoptotic state was detected but with no change in apoptosis rates as evidenced by DNA fragmentation or positivity for the caspase-cleaved epitope of cytokeratin-18 cytoskeletal protein (M30) [82]. In agreement with that, results from our laboratory showed that aPL moderately increased proliferation of the EVT cell line HTR-8/SVneo with no change in the rate of apoptosis [83]. On the other hand, Mulla and colleagues showed that lower concentrations of aPL also moderately stimulated proliferation of HTR-8/SVneo cells but as aPL concentration increased, viability of trophoblast cells were significantly reduced mediated by increased activity of caspase-8, caspase-9 and caspase-3 [52]. An aPL-induced HTR-8/SVneo cell death was at least partially due to increased proinflammatory response of HTR-8/SVneo cells to aPL treatment [52]. Discrepancy between results of different studies on the extent of aPL-induced effects on trophoblast cell survival are most probably due to the heterogeneity of aPL used in experiments as well as the duration of treatment.
Taken together, these observations suggest that apoptosis might be an important mechanism in aPL-induced defective placentation in OAPS, without necessarily involving thrombotic phenomena.

3.2. Trophoblast Cell Function

Along its invasive pathway, trophoblast undergoing EMT acquire markers of an invasive phenotype—integrins α5β1 and α1β1 (forming fibronectin and laminin/collagen receptors), among others, and start to secrete proteolytic enzymes, of which matrix metalloproteinases (MMP) -2 and -9 have the most important role [84,85]. A subpopulation of EVTs that invades the spiral arteries, designated endovascular trophoblast, has been shown to express specific endothelial markers, such as integrin αVβ3, VE-cadherin and α4 integrins [85,86].
A number of in vitro studies on trophoblast cells showed that aPL directly alter trophoblast cell function including invasion and migration abilities. IgG isolated from patients with APS as well as monoclonal antibodies reactive with β2GPI suppressed trophoblast invasion, as shown using several trophoblast cell models including primary trophoblast isolated from first or third trimester placenta, as well as normal trophoblast HTR-8/SVneo and choriocarcinoma JAR cell lines [65,66,67,68,69]. Moreover, Poulton et al. showed that IgG purified from OAPS patients but not from vascular APS patients inhibited HTR-8/SVneo invasion expressing the difference between these two APS subtypes [87].
Further investigations showed that aPL-associated reduction in invasiveness could at least, in part, be attributed to downregulation in invasion mediators—integrin subunits α1, α5 and β1 [67,68,88,89]. MMP-2 and MMP-9 are highly expressed during implantation and early stages of pregnancy, playing a key role in the degradation of the extracellular matrix (ECM) by trophoblast cells [90,91]. Anti-β2GPI antibodies inhibited MMP-2 and MMP-9 secretion by trophoblast cells [66,68], while IgG from aPL-positive sera (aPL IgG) significantly decreased the level of MMP-9 and the overall gelatinolytic capacity of HTR-8/SVneo cells as assessed by in situ gelatin zymography [89]. Another possible mechanism of aPL-mediated invasion suppression is through downregulation of IL-6 secretion and STAT3 activity, as this was shown for mouse anti-β2GPI antibodies in HTR-8/SVneo cells [56]. This cytokine acts as an important regulator of the implantation and placentation processes [56]. Both mouse anti-β2GPI and patient-derived aPL induced an inflammatory response in trophoblast cells, which may result in a negative impact on trophoblast cell function, including an invasive capacity [52,92,93,94].
Our previous findings further support the hypothesis of the direct negative effect of aPL on trophoblast invasion process. Galectin-1 (Gal-1), lectin abundantly present at the feto–maternal interface, is an important part of the trophoblast invasion machinery, as it modulates trophoblast adhesive and invasive capacities [95]. This lectin has the ability to bind various ECM proteins, as well as cell surface adhesion molecules, including trophoblast integrin β1 [96]. Results obtained in our previous research on HTR-8/SVneo cells showed that Gal-1 was reduced in conditioned media of aPL IgG-treated cells, while total cell protein remained unaltered, suggesting that aPL IgG may affect Gal-1 secretion in a manner not yet elucidated [89]. We have shown that aPL IgG treatment activates the p38 MAPK signaling pathway, and that inhibitory effects on integrin subunits and secreted Gal-1 were dependent on this activation [89]. The role of the p38 MAPK signaling pathway was shown in multiple processes involving trophoblast, such as the stimulation of trophoblast cell motility by EGF, demonstrated in experiments on EVT cell line SGHPL-4 [97].
Furthermore, aPL seem to interfere with the trophoblast ability to differentiate into an endothelial-like phenotype contributing to aPL-associated aberrant spiral artery remodeling. Placentas of mice treated with anti-β2GPI antibodies during gestation showed a number of pathological changes including defective vascular remodeling [69]. Moreover, in vitro experiments detected that anti-β2GPI antibodies inhibited the HTR-8/SVneo tube formation ability [69] and compromised the trophoblast ability to bind and integrate into the endothelium [98]. Previous research showed that treatment of primary term trophoblast with aPL IgG downregulated VE-cadherin expression [88], an adhesion molecule important for trophoblast–endothelial interaction, endovascular invasion and spiral artery remodeling [99,100]. Moreover, integrin subunit α4 was also decreased by the treatment [89]. This integrin subunit is upregulated in endovascular trophoblast, probably as one of the adhesion molecules needed to facilitate adhesion to vascular epithelia, where α4β1 integrin is proposed to bind the vascular cell adhesion molecule-1 (VCAM-1) [86]. The schematic representation of the possible mechanisms of aPL action at the site of implantation, based on in vitro findings is given in Figure 1B.
Recently, Yes-associated protein (YAP), the transcription co-activator of the Hippo signaling pathway, was proposed to act as key effector molecule which links aPL-induced upstream intracellular signals and alteration of different trophoblast functions [101]. Namely, it was detected that aPL treatment decreased YAP protein levels in HTR-8/SVneo cells [101]. YAP downregulation increased apoptosis, inhibited migration, invasion and tube formation ability of HTR-8/SVneo cells [101,102,103]. Previous studies of early implantation and trophoblast development showed that inhibition of YAP reduced endometrial attachment, outgrowth, and trophoblast gene expressions of human embryonic stem cell–derived trophoblastic spheroids [104]. Furthermore, decreased YAP expression levels were found in PE placentas [102,103]. All the data present the importance of YAP regulation of trophoblast function, indicating the diversity of effects aPL-induced YAP downregulation could have in establishment and maintenance of pregnancy.
In conclusion, the findings of our and other groups’ research suggest that aPL may induce defective placentation by reducing trophoblast invasion through inhibiting effector molecules—integrins and MMPs and by limiting the amount of Gal-1 present extracellularly. Furthermore, these autoantibodies may interfere with the modification of spiral arteries through downregulation of adhesion molecules characteristic for endovascular trophoblast. Other mechanisms are not excluded.

3.3. Internalization of aPL in Trophoblast Cells

In addition to cell invasion, other cell functions specific for trophoblast have also been shown to be negatively influenced by aPL. Cytotrophoblast fusion and production of a major trophoblast-derived hormone βhCG, necessary for pregnancy progression, were suppressed by aPL [54,105,106,107]. Additionally, a recent study linked aPL to syncitiotrophoblast oxidative stress [108]. This is in accordance with the high heterogeneity of these antibodies. What is still not well understood is the exact mechanism through which these autoantibodies initiate these cellular responses, but both cell surface and intracellular antigens have been proposed as targets.
The ability of patient-derived autoantibodies to penetrate living cells was first proposed more than 40 years ago [109]. Since then, this property was shown for a number of autoantibodies, which led to the conclusion that intracellular antigens are not immunologically privileged, as first thought to be. Several studies have demonstrated that different types of aPL may be detected intracellularly. A study by Galve-de Rochemonteix and colleagues (2000) showed that aPL are accumulated in late endosomes of baby hamster kidney (BHK) cells in culture [110]. A more recent study demonstrated that anti-phosphatidylethanolamine antibodies target the cytosolic surface of early endosomes in human umbilical vein endothelial cells (HUVECs) and hypothesizes that these antibodies could have important implications for a wide range of biological processes in different cell types [111]. Trying to elucidate the mechanism of interaction between anti–β2GPI monoclonal antibodies WB-6 with resting monocytes, Virachith and colleagues (2019) found that WB-6 exhibits binding activity to DNA and enters living monocytes [112].
It is well known that the passive immunity provided to the human fetus is in part obtained through the transport of maternal IgG across the syncytiotrophoblast. Even though the transport of maternal IgG increases after the 22nd week of gestation and is mediated by the Fcγ receptor, there are data indicating that transport across early placental trophoblast is not limited by a lack of specific IgG receptors. Furthermore, aPL have been shown to bind EVTs [68,113,114], and also to bind and internalize into syncytiotrophoblast [60]. This process was independent on Fc-receptor, as shown by employment of anti-β2GPI that lacks Fc fragment but was dependent on the low-density lipoprotein receptor (LDLR) [60]. Extrusion of necrotic trophoblast debris from syncytiotrophoblast, caused by aPL treatment, was dependent on the internalization of these antibodies [60]. A subsequent study from the same group showed that the same aPL are not internalized in the EVTs from explanted villi [114]. Our study, however, demonstrated that patient-derived aPL IgG was able to enter and accumulate in the primary first trimester cytotrophoblast in culture and HTR-8/SVneo cells in a time-dependent manner [113], similarly to findings of Hou and colleagues on HUVECs [111]. We suspect that this discrepancy could be due to the longer exposure time in our approach and, to a lesser degree, to the difference in species of origin and specificity of the used antibodies.
Based on the collected data from studies on the aPL potential to enter trophoblast as well as other types of cells, it can be concluded that there is a whole spectra of possible cell surface as well as intracellular targets for these autoantibodies, implicating other possible routes for aPL’s influence on trophoblast cell function.

4. Non-Coding RNAs—Emerging Players in OAPS Pathophysiology

Being multifunctional regulators of different biological processes, as the growing body of evidence suggests, it is not surprising that there is increasing interest in studying the involvement of non-coding RNAs (ncRNAs) in the pathophysiology of APS and other autoimmune disorders [25,115,116,117,118,119,120,121]. NcRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), regulate gene expression on transcriptional and post-transcriptional levels [122,123]. Aberrant expression of ncRNAs could significantly affect various cellular processes eventually leading to the development of different pathologies [124,125,126].
An in vitro study on trophoblast HTR-8/SVneo cells revealed that anti-β2GPI upregulated cellular and exosome levels of miR-146a-5p, miR-146a-3p, miR-155 and miR-210 [93]. Moreover, women positive for aPL experiencing pregnancy complications had elevated circulating miR-146a-3p levels compared to healthy controls [93]. Previous research showed that aPL stimulated a proinflammatory response in trophoblast cells [52]. Specifically, anti-β2GPI elicited IL-8 secretion among other cytokines in HTR-8/SVneo cells through the TLR4/MyD88 pathway [52]. Since miR-146a-5p, miR-155 and miR-210 were shown to mediate TLR signaling [127,128,129,130], Gysler and colleagues investigated the role of these miRNAs in TLR4-dependent IL-8 secretion in aPL-treated trophoblast cells [93]. They showed that aPL-stimulated upregulation of miR-146a-5p, miR-146a-3p and miR-210 but not miR-155 was TLR4 dependent. However, anti-β2GPI-induced IL-8 secretion was shown to be mediated by miR-146a-3p and it was through the activation of RNA sensor TLR8 [93].
MiR-146a, miR-155 and miR-210 are important regulators of numerous cellular processes. Although there are some studies with opposing results [131,132], a number of studies showed that overexpression of miR-146a [133,134,135,136], miR-155 [137,138,139] and miR-210 [140,141,142,143] inhibited migration and invasion of trophoblast cells. MiR-146a and miR-155 are major regulators of the immune response and disrupted expression of these miRNAs has been associated with pathologies characterized by chronic inflammation [144]. MiR-146a affects trophoblast EMT, migration and invasion abilities through direct downregulation of TNF receptor-associated factor 6 (TRAF6) [134] and atypical chemokine receptor 2 (ACKR2) [136]. TRAF6 is a signal transducer in the TLR4/MyD88 pathway involved in the regulation of the immune response [145,146] but also implicated in different cellular processes including regulation of proliferation, migration and invasion of cancer cells [146]. ACKR2, a chemokine scavenger, is involved in the maintenance of balance between pro and anti-inflammatory cytokines at the feto–maternal interface [147] and dysregulation of this molecule is associated with different pregnancy complications [148,149]. Wnt/β-catenin signaling pathway was also found to be affected by miR-146a-5p through direct regulation of Wnt2 expression [135]. Furthermore, it was shown that overexpression of miR-146a downregulated CXCR4 and EGFR in HTR-8/SVneo cells [133] affecting signaling pathways activated through these two receptors which are involved in regulation of trophoblast migration and invasion [150,151]. MiR-155 affects the TGF-β/Smad signaling pathway important for regulation of EVT invasion [152] directly targeting expression of Smad2 [153]. Other direct miR-155 targets were also implicated in the regulation of trophoblast cell functions including angiogenic factor CYR61 [137], an important regulator of cell cycle progression cyclin D1 [154], eNOS [139] and forkhead-box class O transcription factor 3 (FOXO3) [155].
MiR-210 is a master hypoxamiR, a miRNA whose expression is induced by hypoxic conditions [156]. It is an important regulator of mitochondrial metabolism, cell proliferation and differentiation, angiogenesis and other oxygen-sensitive processes [156]. Overexpression of miR-210 in trophoblast cells inhibited mitochondrial respiration which consequently could lead to generation of excessive amounts of reactive oxygen species and increased placental oxidative stress [157]. MiR-210 inhibited HTR-8/SVneo invasion via ERK/MAPK-dependent mechanism [140]. Overexpression of miR-210 dysregulated expression of EMT-related proteins and consequentially inhibited invasive abilities of trophoblast cells [143]. Namely, expression of the mesenchymal marker vimentin and N-cadherin, a promoter of EMT, was decreased while E-cadherin, an epithelial marker, was upregulated in HTR-8/SVneo cells overexpressing miR-210 [143]. Moreover, trophoblast upregulation of miR-210-3p has been associated with impaired remodeling of spiral arteries [142]. In this study, authors showed that miR-210-3p-dependent impairment of trophoblast function is mediated through direct dysregulation of caudal-related homeobox transcription factor 2 (CDX2), essential transcription factor for trophoblast differentiation active during blastocyst development [142].
Reduced trophoblast invasion and spiral artery remodeling are associated with pregnancy complications characteristic for OAPS patients, such as RPL and PE [14,17]. Since increased levels of miR-146a, miR-155 and miR-210 have been found in placentas of aPL-negative RPL [134,158,159,160] and PE patients [135,136,137,139,161,162,163] it could be concluded that aPL-induced upregulation of named miRNAs in trophoblast cells is a contributing factor to the development of these obstetric complications in OAPS patients. Moreover, increased levels of miR-146a, miR-155 and miR-210 were found in blood of patients suffering from pregnancy loss and/or PE [140,164,165,166,167] suggesting the use of these miRNAs as biomarkers for early diagnostics as well as mechanism-based targets of new therapeutics for RPL and PE associated or not with OAPS.
As mentioned above, APS is one of the main risk factors for RPL [168] but underlying mechanisms of aPL-induced RPL are still not completely elucidated. Recent studies identified lncRNA MALAT1 as one of the major regulators of the processes important for the adequate placental development and function in early pregnancy [158,169,170,171,172,173]. Namely, MALAT1 levels were significantly downregulated in placentas of the patients experiencing RPL of unknown etiology compared to the healthy controls [158,174]. Furthermore, recent research showed that trophoblast and placental MALAT1 levels of aPL-positive RPL patients were even lower than MALAT1 levels of aPL-negative RPL patients [172]. Furthermore, aPL-positive RPL mouse model was generated [172]. The embryo resorption rate was increased in aPL-positive RPL mice compared both to RPL mice negative for aPL and control mice [172]. Placental MALAT1 overexpression by adenoviral transfection in aPL-positive RPL mice significantly decreased embryo resorption rate compared to untreated aPL-RPL mice [172].
Moreover, low expression levels of MALAT1 were also found in placentas of PE patients comparing with normal controls [170,171,173,175]. Effects of decreased MALAT1 expression on trophoblast cell function were investigated in vitro by downregulation of this lncRNA in HTR-8/SVneo and JAR cells. The results showed that trophoblast cells with decreased MALAT1 levels proliferated less than unmodified cells and their migrating and invasive abilities were decreased as well as expression of EMT-related proteins [158,169,170,171,172,173]. Different mechanisms of action were proposed for MALAT1-dependent regulation of trophoblast cell function. It was shown that MALAT1 modulates IGF-1/PI3K/Akt signaling affecting trophoblast migration and invasion abilities [169]. Furthermore, VEGFA was found to be a downstream mediator of MALAT1-depepndent inhibition of trophoblast proliferation [173] and endovascular differentiation [171].
Among other mechanisms of action, lncRNAs can regulate gene expression by affecting miRNA expression and activity via sequestration [123,176]. Some lncRNAs, part of competing endogenous RNA (ceRNA) family, act as molecular miRNA sponges. They competitively bind specific miRNAs and thus prevent them from binding to their target mRNAs. In that way, negative miRNA effect on target gene expression is reduced [123,176]. Mutual regulation of miRNA and lncRNA activities is involved in regulation and fine tuning of many biological processes. Growing body of evidence has shown that MALAT1 functions as ceRNA [158,169,173,177,178,179,180,181,182]. Among other miRNAs, miR-146a [158,177,178,179] and miR-155 [183,184] have also been shown to be MALAT1 binding partners. These studies indicate the involvement of MALAT1/miR-146a and/or miR-155 regulatory axes in diverse negative effects of aPL on placental function leading to RPL or other obstetrical complications in OAPS.
Recently, another lncRNA named LncNR_040117 has been identified as an important mediator of APS-induced RPL [24]. Firstly, it has been shown that platelet-derived microparticles (PMPs) isolated during first trimester of pregnancy from APS patients with the history of RPL stimulated apoptosis and inhibited invasion and migration of trophoblast HTR-8/SVneo cells [185]. PMPs are vesicles derived from platelets undergone activation or apoptosis [186]. They are the most abundant type of microparticles present in human circulation and they were associated with different pathologies such as cancer, cardiovascular and autoimmune diseases [186,187,188]. PMPs mediate various physiological processes affecting target cells thorough specific interactions including surface receptor signaling and delivering of bioactive molecules such as cytokines, enzymes, growth factors and RNAs [186,187,188,189,190]. Content of PMPs’ cargo molecules depend on the signals activating platelets and stimulating PMPs’ formation and it is modified in pathological conditions [186,187,188]. lncRNA profiling of APS-associated PMPs identified LncNR_040117 as one of the significantly overexpressed lncRNAs in PMPs isolated from APS patients with the history of RPL comparing to the gestational age matched healthy controls [24]. Moreover, this lncRNA has been proposed for biomarker of APS-induced RPL [24]. Downregulation of LncNR_040117 stimulated proliferation, migration and invasion of transfected HTR-8/SVneo cells [24,191]. These results indicate that LncNR_040117 upregulation by intake of APS-related PMPs could have opposite, detrimental effects on trophoblast function. On the other hand, finding strategies for targeted blocking of this lncRNA could be a potential method of preventing miscarriage in APS patients. The possible implications of APS-associated ncRNAs in trophoblast cell function are summarized in Table 2.
NcRNA research field is constantly developing, giving insights in complex regulation of biological processes in health and disease. Future elucidation of various participants in these fine-tuned processes will provide new opportunities for the development of potential therapeutics and strategies for management of obstetrical complications related to APS.

5. Conclusions

Given that APS still represents one of the most common threats for pregnancy complications, the current knowledge regarding placental dysfunction in APS must be significantly improved. There are multiple possible mechanisms involved in APS-associated placental dysfunction. According to the evidence from in vitro and in vivo studies, both extracellular and intracellular antigens may be targeted by aPL, activating different cellular responses that further cause excessive apoptosis and impaired trophoblast invasion/placentation. The aPL-induced modulation of epigenetic mechanisms such as changing ncRNAs expression is emerging as a key contributor to APS progression.
Current treatment strategies are not effective for all patients. Conventional treatment strategies mostly include antithrombotic agents, while immunosuppressive therapy has been increasingly used. Non-coding RNAs are emerging players in the pathogenesis of APS. Given that these molecules have been proposed as biomarkers of many pathological conditions and as therapeutic targets, future investigations could be directed towards identification of differentially expressed ncRNAs in APS, and elucidation of their roles in the pathogenesis of APS. This would provide basis for the development of ncRNA-targeting treatments. Several ncRNA-targeting drugs for other conditions are already being tested in clinical trials.
Taken together, further investigations are needed to fully understand the causes of APS-associated pregnancy complications, so that every affected woman can be treated adequately.

Author Contributions

Conceptualization, writing—review and editing, S.V.P.; writing—original draft preparation, review and editing, A.V.; writing—original draft preparation, review and editing, Ž.B.-T.; writing—original draft preparation, S.K.; writing—original draft preparation, M.P.; writing—original draft preparation, review and editing, L.V.; conceptualization, writing—original draft preparation, review and editing, visualization, M.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Science, Technological Development and Innovation of Serbia, contract number 451-03-47/2023-01/200019.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Schreiber, K.; Sciascia, S.; de Groot, P.G.; Devreese, K.; Jacobsen, S.; Ruiz-Irastorza, G.; Salmon, J.E.; Shoenfeld, Y.; Shovman, O.; Hunt, B.J. Antiphospholipid Syndrome. Nat. Rev. Dis. Prim. 2018, 4, 17103. [Google Scholar] [CrossRef] [PubMed]
  2. Knight, J.S.; Branch, D.W.; Ortel, T.L. Antiphospholipid Syndrome: Advances in Diagnosis, Pathogenesis, and Management. BMJ 2023, 380, e069717. [Google Scholar] [CrossRef] [PubMed]
  3. Miyakis, S.; Lockshin, M.D.; Atsumi, T.; Branch, D.W.; Brey, R.L.; Cervera, R.; Derksen, R.H.W.M.; De Groot, P.G.; Koike, T.; Meroni, P.L.; et al. International Consensus Statement on an Update of the Classification Criteria for Definite Antiphospholipid Syndrome (APS). J. Thromb. Haemost. 2006, 4, 295–306. [Google Scholar] [CrossRef] [PubMed]
  4. Pons-Estel, G.J.; Andreoli, L.; Scanzi, F.; Cervera, R.; Tincani, A. The Antiphospholipid Syndrome in Patients with Systemic Lupus Erythematosus. J. Autoimmun. 2017, 76, 10–20. [Google Scholar] [CrossRef]
  5. Cervera, R.; Khamashta, M.A.; Shoenfeld, Y.; Camps, M.T.; Jacobsen, S.; Kiss, E.; Zeher, M.M.; Tincani, A.; Kontopoulou-Griva, I.; Galeazzi, M.; et al. Morbidity and Mortality in the Antiphospholipid Syndrome during a 5-Year Period: A Multicentre Prospective Study of 1000 Patients. Ann. Rheum. Dis. 2009, 68, 1428–1432. [Google Scholar] [CrossRef]
  6. Rodziewicz, M.; D’Cruz, D.P.; Gulliford, M. O30 The Epidemiology of Antiphospholipid Syndrome in the UK, 1990–2016. Rheumatology 2019, 58, kez105-029. [Google Scholar] [CrossRef]
  7. Duarte-García, A.; Pham, M.M.; Crowson, C.S.; Amin, S.; Moder, K.G.; Pruthi, R.K.; Warrington, K.J.; Matteson, E.L. The Epidemiology of Antiphospholipid Syndrome: A Population-Based Study. Arthritis Rheumatol. 2019, 71, 1545–1552. [Google Scholar] [CrossRef]
  8. Hwang, J.J.; Shin, S.H.; Kim, Y.J.; Oh, Y.M.; Lee, S.D.; Kim, Y.H.; Choi, C.W.; Lee, J.S. Epidemiology of Antiphospholipid Syndrome in Korea: A Nationwide Population-Based Study. J. Korean Med. Sci. 2020, 35, e35. [Google Scholar] [CrossRef]
  9. Dabit, J.Y.; Valenzuela-Almada, M.O.; Vallejo-Ramos, S.; Duarte-García, A. Epidemiology of Antiphospholipid Syndrome in the General Population. Curr. Rheumatol. Rep. 2022, 23, 85. [Google Scholar] [CrossRef]
  10. Cervera, R.; Piette, J.-C.; Font, J.; Khamashta, M.A.; Shoenfeld, Y.; Camps, M.T.; Jacobsen, S.; Lakos, G.; Tincani, A.; Kontopoulou-Griva, I.; et al. Antiphospholipid Syndrome: Clinical and Immunologic Manifestations and Patterns of Disease Expression in a Cohort of 1000 Patients. Arthritis Rheum. 2002, 46, 1019–1027. [Google Scholar] [CrossRef]
  11. Ruiz-Irastorza, G.; Egurbide, M.-V.; Ugalde, J.; Aguirre, C. High Impact of Antiphospholipid Syndrome on Irreversible Organ Damage and Survival of Patients with Systemic Lupus Erythematosus. Arch. Intern. Med. 2004, 164, 77–82. [Google Scholar] [CrossRef] [PubMed]
  12. Meroni, P.L.; Borghi, M.O.; Grossi, C.; Chighizola, C.B.; Durigutto, P.; Tedesco, F. Obstetric and Vascular Antiphospholipid Syndrome: Same Antibodies but Different Diseases? Nat. Rev. Rheumatol. 2018, 14, 433–440. [Google Scholar] [CrossRef] [PubMed]
  13. Taraborelli, M.; Reggia, R.; Dall’Ara, F.; Fredi, M.; Andreoli, L.; Gerosa, M.; Hoxha, A.; Massaro, L.; Tonello, M.; Costedoat-Chalumeau, N.; et al. Longterm Outcome of Patients with Primary Antiphospholipid Syndrome: A Retrospective Multicenter Study. J. Rheumatol. 2017, 44, 1165–1172. [Google Scholar] [CrossRef] [PubMed]
  14. Alijotas-Reig, J.; Esteve-Valverde, E.; Anunciación-Llunell, A.; Marques-Soares, J.; Pardos-Gea, J.; Miró-Mur, F. Pathogenesis, Diagnosis and Management of Obstetric Antiphospholipid Syndrome: A Comprehensive Review. J. Clin. Med. 2022, 11, 675. [Google Scholar] [CrossRef] [PubMed]
  15. Niznik, S.; Rapoport, M.J.; Avnery, O.; Lubetsky, A.; Shavit, R.; Ellis, M.H.; Agmon-Levin, N. Long Term Follow up of Patients with Primary Obstetric Antiphospholipid Syndrome. Front. Pharmacol. 2022, 13, 824775. [Google Scholar] [CrossRef]
  16. Ripoll, V.M.; Pregnolato, F.; Mazza, S.; Bodio, C.; Grossi, C.; McDonnell, T.; Pericleous, C.; Meroni, P.L.; Isenberg, D.A.; Rahman, A.; et al. Gene Expression Profiling Identifies Distinct Molecular Signatures in Thrombotic and Obstetric Antiphospholipid Syndrome. J. Autoimmun. 2018, 93, 114–123. [Google Scholar] [CrossRef]
  17. Alijotas-Reig, J.; Esteve-Valverde, E.; Ferrer-Oliveras, R.; Sáez-Comet, L.; Lefkou, E.; Mekinian, A.; Belizna, C.; Ruffatti, A.; Tincani, A.; Marozio, L.; et al. The European Registry on Obstetric Antiphospholipid Syndrome (EUROAPS): A Survey of 1000 Consecutive Cases. Autoimmun. Rev. 2019, 18, 406–414. [Google Scholar] [CrossRef]
  18. Erton, Z.B.; Sevim, E.; de Jesús, G.R.; Cervera, R.; Ji, L.; Pengo, V.; Ugarte, A.; Andrade, D.; Andreoli, L.; Atsumi, T.; et al. Pregnancy Outcomes in Antiphospholipid Antibody Positive Patients: Prospective Results from the AntiPhospholipid Syndrome Alliance for Clinical Trials and InternatiOnal Networking (APS ACTION) Clinical Database and Repository ('Registry’). Lupus Sci. Med. 2022, 9, e000633. [Google Scholar] [CrossRef]
  19. Cervera, R. Antiphospholipid Syndrome. Thromb. Res. 2017, 151 (Suppl. S1), S43–S47. [Google Scholar] [CrossRef]
  20. Cohen, H.; Cuadrado, M.J.; Erkan, D.; Duarte-Garcia, A.; Isenberg, D.A.; Knight, J.S.; Ortel, T.L.; Rahman, A.; Salmon, J.E.; Tektonidou, M.G.; et al. 16th International Congress on Antiphospholipid Antibodies Task Force Report on Antiphospholipid Syndrome Treatment Trends. Lupus 2020, 29, 1571–1593. [Google Scholar] [CrossRef]
  21. Sammaritano, L.R.; Bermas, B.L.; Chakravarty, E.E.; Chambers, C.; Clowse, M.E.B.; Lockshin, M.D.; Marder, W.; Guyatt, G.; Branch, D.W.; Buyon, J.; et al. 2020 American College of Rheumatology Guideline for the Management of Reproductive Health in Rheumatic and Musculoskeletal Diseases. Arthritis Rheumatol. 2020, 72, 529–556. [Google Scholar] [CrossRef]
  22. Alijotas-Reig, J.; Esteve-Valverde, E.; Llurba, E.; Gris, J.M. Treatment of Refractory Poor APL-Related Obstetric Outcomes with TNF-Alpha Blockers: Maternal-Fetal Outcomes in a Series of 18 Cases. Semin. Arthritis Rheum. 2019, 49, 314–318. [Google Scholar] [CrossRef] [PubMed]
  23. Salmon, J.E.; Guerra, M.; Kim, M.; Branch, D.W. 1201 IMPACT Study: Preliminary Results of a Trial with a Biologic to Prevent Preeclampsia in Women with Antiphospholipid Syndrome. Lupus Sci. Med. 2022, 9, A84. [Google Scholar] [CrossRef]
  24. Sun, Z.; Zhou, Q.; Yang, Y.; Li, L.; Yu, M.; Li, H.; Li, A.; Wang, X.; Jiang, Y. Identification and Ultrasensitive Photoelectrochemical Detection of LncNR_040117: A Biomarker of Recurrent Miscarriage and Antiphospholipid Antibody Syndrome in Platelet-Derived Microparticles. J. Nanobiotechnol. 2022, 20, 396. [Google Scholar] [CrossRef] [PubMed]
  25. Tan, Y.; Liu, Q.; Li, Z.; Yang, S.; Cui, L. Epigenetics-Mediated Pathological Alternations and Their Potential in Antiphospholipid Syndrome Diagnosis and Therapy. Autoimmun. Rev. 2022, 21, 103130. [Google Scholar] [CrossRef]
  26. Misasi, R.; Longo, A.; Recalchi, S.; Caissutti, D.; Riitano, G.; Manganelli, V.; Garofalo, T.; Sorice, M.; Capozzi, A. Molecular Mechanisms of “Antiphospholipid Antibodies” and Their Paradoxical Role in the Pathogenesis of “Seronegative APS”. Int. J. Mol. Sci. 2020, 21, 8411. [Google Scholar] [CrossRef]
  27. Kaburaki, J.; Kuwana, M.; Yamamoto, M.; Kawai, S.; Ikeda, Y. Clinical Significance of Anti-Annexin V Antibodies in Patients with Systemic Lupus Erythematosus. Am. J. Hematol. 1997, 54, 209–213. [Google Scholar] [CrossRef]
  28. Salle, V.; Mazière, J.C.; Smail, A.; Cévallos, R.; Mazière, C.; Fuentes, V.; Tramier, B.; Makdassi, R.; Choukroun, G.; Vittecoq, O.; et al. Anti-Annexin II Antibodies in Systemic Autoimmune Diseases and Antiphospholipid Syndrome. J. Clin. Immunol. 2008, 28, 291–297. [Google Scholar] [CrossRef]
  29. Oosting, J.D.; Derksen, R.H.; Bobbink, I.W.; Hackeng, T.M.; Bouma, B.N.; de Groot, P.G. Antiphospholipid Antibodies Directed against a Combination of Phospholipids with Prothrombin, Protein C, or Protein S: An Explanation for Their Pathogenic Mechanism? Blood 1993, 81, 2618–2625. [Google Scholar] [CrossRef]
  30. Sorice, M.; Arcieri, P.; Griggi, T.; Circella, A.; Misasi, R.; Lenti, L.; Di Nucci, G.D.; Mariani, G. Inhibition of Protein S by Autoantibodies in Patients with Acquired Protein S Deficiency. Thromb. Haemost. 1996, 75, 555–559. [Google Scholar] [CrossRef]
  31. Sanmarco, M.; Gayet, S.; Alessi, M.-C.; Audrain, M.; de Maistre, E.; Gris, J.-C.; de Groot, P.G.; Hachulla, E.; Harlé, J.-R.; Sié, P.; et al. Antiphosphatidylethanolamine Antibodies Are Associated with an Increased Odds Ratio for Thrombosis. A Multicenter Study with the Participation of the European Forum on Antiphospholipid Antibodies. Thromb. Haemost. 2007, 97, 949–954. [Google Scholar] [PubMed]
  32. Alessandri, C.; Bombardieri, M.; Di Prospero, L.; Conigliaro, P.; Conti, F.; Labbadia, G.; Misasi, R.; Sorice, M.; Valesini, G. Anti-Lysobisphosphatidic Acid Antibodies in Patients with Antiphospholipid Syndrome and Systemic Lupus Erythematosus. Clin. Exp. Immunol. 2005, 140, 173–180. [Google Scholar] [CrossRef] [PubMed]
  33. Arvieux, J.; Darnige, L.; Caron, C.; Reber, G.; Bensa, J.C.; Colomb, M.G. Development of an ELISA for Autoantibodies to Prothrombin Showing Their Prevalence in Patients with Lupus Anticoagulants. Thromb. Haemost. 1995, 74, 1120–1125. [Google Scholar] [CrossRef]
  34. Sciascia, S.; Sanna, G.; Murru, V.; Roccatello, D.; Khamashta, M.A.; Bertolaccini, M.L. Anti-Prothrombin (APT) and Anti-Phosphatidylserine/Prothrombin (APS/PT) Antibodies and the Risk of Thrombosis in the Antiphospholipid Syndrome. A Systematic Review. Thromb. Haemost. 2014, 111, 354–364. [Google Scholar] [CrossRef] [PubMed]
  35. Ortona, E.; Capozzi, A.; Colasanti, T.; Conti, F.; Alessandri, C.; Longo, A.; Garofalo, T.; Margutti, P.; Misasi, R.; Khamashta, M.A.; et al. Vimentin/Cardiolipin Complex as a New Antigenic Target of the Antiphospholipid Syndrome. Blood 2010, 116, 2960–2967. [Google Scholar] [CrossRef]
  36. Andreoli, L.; Chighizola, C.B.; Banzato, A.; Pons-Estel, G.J.; De Jesus, G.R.; Erkan, D. Estimated Frequency of Antiphospholipid Antibodies in Patients with Pregnancy Morbidity, Stroke, Myocardial Infarction, and Deep Vein Thrombosis: A Critical Review of the Literature. Arthritis Care Res. 2013, 65, 1869–1873. [Google Scholar] [CrossRef]
  37. Foddai, S.G.; Radin, M.; Cecchi, I.; Gaito, S.; Orpheu, G.; Rubini, E.; Barinotti, A.; Menegatti, E.; Mengozzi, G.; Roccatello, D.; et al. The Prevalence of Antiphospholipid Antibodies in Women with Late Pregnancy Complications and Low-Risk for Chromosomal Abnormalities. J. Thromb. Haemost. 2020, 18, 2921–2928. [Google Scholar] [CrossRef]
  38. Rai, R.; Regan, L. Antiphospholipid Syndrome and Pregnancy Loss. Hosp. Med. 1998, 59, 637–639. [Google Scholar]
  39. Page, J.M.; Christiansen-Lindquist, L.; Thorsten, V.; Parker, C.B.; Reddy, U.M.; Dudley, D.J.; Saade, G.R.; Coustan, D.; Rowland Hogue, C.J.; Conway, D.; et al. Diagnostic Tests for Evaluation of Stillbirth. Obstet. Gynecol. 2017, 129, 699–706. [Google Scholar] [CrossRef]
  40. Gkrouzman, E.; Sevim, E.; Finik, J.; Andrade, D.; Pengo, V.; Sciascia, S.; Tektonidou, M.G.; Ugarte, A.; Chighizola, C.B.; Belmont, H.M.; et al. Antiphospholipid Antibody Profile Stability Over Time: Prospective Results From the APS ACTION Clinical Database and Repository. J. Rheumatol. 2021, 48, 541–547. [Google Scholar] [CrossRef]
  41. Hughes, G.R.V.; Khamashta, M.A. ‘Seronegative Antiphospholipid Syndrome’: An Update. Lupus 2019, 28, 273–274. [Google Scholar] [CrossRef] [PubMed]
  42. Sanmarco, M. Clinical Significance of Antiphosphatidylethanolamine Antibodies in the So-Called “Seronegative Antiphospholipid Syndrome”. Autoimmun. Rev. 2009, 9, 90–92. [Google Scholar] [CrossRef] [PubMed]
  43. Abisror, N.; Nguyen, Y.; Marozio, L.; Esteve Valverde, E.; Udry, S.; Pleguezuelo, D.E.; Billoir, P.; Mayer-Pickel, K.; Urbanski, G.; Zigon, P.; et al. Obstetrical Outcome and Treatments in Seronegative Primary APS: Data from European Retrospective Study. RMD open 2020, 6, e001340. [Google Scholar] [CrossRef] [PubMed]
  44. Pires da Rosa, G.; Ferreira, E.; Sousa-Pinto, B.; Rodríguez-Pintó, I.; Brito, I.; Mota, A.; Cervera, R.; Espinosa, G. Comparison of Non-Criteria Antiphospholipid Syndrome with Definite Antiphospholipid Syndrome: A Systematic Review. Front. Immunol. 2022, 13, 967178. [Google Scholar] [CrossRef]
  45. Alijotas-Reig, J.; Esteve-Valverde, E.; Ferrer-Oliveras, R.; Sáez-Comet, L.; Lefkou, E.; Mekinian, A.; Belizna, C.; Ruffatti, A.; Hoxha, A.; Tincani, A.; et al. Comparative Study of Obstetric Antiphospholipid Syndrome (OAPS) and Non-Criteria Obstetric APS (NC-OAPS): Report of 1640 Cases from the EUROAPS Registry. Rheumatology 2020, 59, 1306–1314. [Google Scholar] [CrossRef]
  46. Pierangeli, S.S.; Chen, P.P.; Raschi, E.; Scurati, S.; Grossi, C.; Borghi, M.O.; Palomo, I.; Harris, E.N.; Meroni, P.L. Antiphospholipid Antibodies and the Antiphospholipid Syndrome: Pathogenic Mechanisms. Semin. Thromb. Hemost. 2008, 34, 236–250. [Google Scholar] [CrossRef]
  47. Amengual, O.; Atsumi, T.; Khamashta, M.A.; Hughes, G.R. The Role of the Tissue Factor Pathway in the Hypercoagulable State in Patients with the Antiphospholipid Syndrome. Thromb. Haemost. 1998, 79, 276–281. [Google Scholar] [CrossRef]
  48. Rand, J.H.; Wu, X.-X.; Quinn, A.S.; Ashton, A.W.; Chen, P.P.; Hathcock, J.J.; Andree, H.A.M.; Taatjes, D.J. Hydroxychloroquine Protects the Annexin A5 Anticoagulant Shield from Disruption by Antiphospholipid Antibodies: Evidence for a Novel Effect for an Old Antimalarial Drug. Blood 2010, 115, 2292–2299. [Google Scholar] [CrossRef]
  49. Chamley, L.W.; Allen, J.L.; Johnson, P.M. Synthesis of Beta2 Glycoprotein 1 by the Human Placenta. Placenta 1997, 18, 403–410. [Google Scholar] [CrossRef]
  50. Burton, G.J.; Woods, A.W.; Jauniaux, E.; Kingdom, J.C.P. Rheological and Physiological Consequences of Conversion of the Maternal Spiral Arteries for Uteroplacental Blood Flow during Human Pregnancy. Placenta 2009, 30, 473–482. [Google Scholar] [CrossRef]
  51. Rand, J.H.; Wu, X.X.; Andree, H.A.; Lockwood, C.J.; Guller, S.; Scher, J.; Harpel, P.C. Pregnancy Loss in the Antiphospholipid-Antibody Syndrome—A Possible Thrombogenic Mechanism. N. Engl. J. Med. 1997, 337, 154–160. [Google Scholar] [CrossRef] [PubMed]
  52. Mulla, M.J.; Brosens, J.J.; Chamley, L.W.; Giles, I.; Pericleous, C.; Rahman, A.; Joyce, S.K.; Panda, B.; Paidas, M.J.; Abrahams, V.M. Antiphospholipid Antibodies Induce a Pro-Inflammatory Response in First Trimester Trophoblast via the TLR4/MyD88 Pathway. Am. J. Reprod. Immunol. 2009, 62, 96–111. [Google Scholar] [CrossRef] [PubMed]
  53. Mulla, M.J.; Weel, I.C.; Potter, J.A.; Gysler, S.M.; Salmon, J.E.; Peraçoli, M.T.S.; Rothlin, C.V.; Chamley, L.W.; Abrahams, V.M. Antiphospholipid Antibodies Inhibit Trophoblast Toll-Like Receptor and Inflammasome Negative Regulators. Arthritis Rheumatol. 2018, 70, 891–902. [Google Scholar] [CrossRef]
  54. Di Simone, N. In Vitro Effect of Antiphospholipid Antibody-Containing Sera on Basal and Gonadotrophin Releasing Hormonedependent Human Chorionic Gonadotrophin Release by Cultured Trophoblast Cells. Placenta 1995, 16, 75–83. [Google Scholar] [CrossRef] [PubMed]
  55. Di Simone, N.; Ferrazzani, S.; Castellani, R.; De Carolis, S.; Mancuso, S.; Caruso, A. Heparin and Low-Dose Aspirin Restore Placental Human Chorionic Gonadotrophin Secretion Abolished by Antiphospholipid Antibody- Containing Sera. Hum. Reprod. 1997, 12, 2061–2065. [Google Scholar] [CrossRef] [PubMed]
  56. Mulla, M.J.; Myrtolli, K.; Brosens, J.J.; Chamley, L.W.; Kwak-Kim, J.Y.; Paidas, M.J.; Abrahams, V.M. Antiphospholipid Antibodies Limit Trophoblast Migration by Reducing IL-6 Production and STAT3 Activity. Am. J. Reprod. Immunol. 2010, 63, 339–348. [Google Scholar] [CrossRef]
  57. Girardi, G.; Berman, J.; Redecha, P.; Spruce, L.; Thurman, J.M.; Kraus, D.; Hollmann, T.J.; Casali, P.; Caroll, M.C.; Wetsel, R.A.; et al. Complement C5a Receptors and Neutrophils Mediate Fetal Injury in the Antiphospholipid Syndrome. J. Clin. Investig. 2003, 112, 1644–1654. [Google Scholar] [CrossRef]
  58. Girardi, G.; Yarilin, D.; Thurman, J.M.; Holers, V.M.; Salmon, J.E. Complement Activation Induces Dysregulation of Angiogenic Factors and Causes Fetal Rejection and Growth Restriction. J. Exp. Med. 2006, 203, 2165–2175. [Google Scholar] [CrossRef]
  59. Shamonki, J.M.; Salmon, J.E.; Hyjek, E.; Baergen, R.N. Excessive Complement Activation Is Associated with Placental Injury in Patients with Antiphospholipid Antibodies. Am. J. Obstet. Gynecol. 2007, 196, 167.e1–167.e5. [Google Scholar] [CrossRef]
  60. Viall, C.A.; Chen, Q.; Liu, B.; Hickey, A.; Snowise, S.; Salmon, J.E.; Stone, P.R.; Chamley, L.W. Antiphospholipid Antibodies Internalised by Human Syncytiotrophoblast Cause Aberrant Cell Death and the Release of Necrotic Trophoblast Debris. J. Autoimmun. 2013, 47, 45–57. [Google Scholar] [CrossRef]
  61. Knöfler, M.; Haider, S.; Saleh, L.; Pollheimer, J.; Gamage, T.K.J.B.; James, J. Human Placenta and Trophoblast Development: Key Molecular Mechanisms and Model Systems. Cell. Mol. Life Sci. 2019, 76, 3479–3496. [Google Scholar] [CrossRef] [PubMed]
  62. Moser, G.; Windsperger, K.; Pollheimer, J.; de Sousa Lopes, S.C.; Huppertz, B. Human Trophoblast Invasion: New and Unexpected Routes and Functions. Histochem. Cell Biol. 2018, 150, 361–370. [Google Scholar] [CrossRef] [PubMed]
  63. Quenby, S.; Mountfield, S.; Cartwright, J.E.; Whitley, G.S.; Chamley, L.; Vince, G. Antiphospholipid Antibodies Prevent Extravillous Trophoblast Differentiation. Fertil. Steril. 2005, 83, 691–698. [Google Scholar] [CrossRef]
  64. Pantham, P.; Abrahams, V.M.; Chamley, L.W. The Role of Anti-Phospholipid Antibodies in Autoimmune Reproductive Failure. Reproduction 2016, 151, R79–R90. [Google Scholar] [CrossRef] [PubMed]
  65. Di Simone, N.; Caliandro, D.; Castellani, R.; Ferrazzani, S.; De Carolis, S.; Caruso, A. Low-Molecular Weight Heparin Restores in-Vitro Trophoblast Invasiveness and Differentiation in Presence of Immunoglobulin G Fractions Obtained from Patients with Antiphospholipid Syndrome. Hum. Reprod. 1999, 14, 489–495. [Google Scholar] [CrossRef]
  66. Blank, M.; Anafi, L.; Zandman-Goddard, G.; Krause, I.; Goldman, S.; Shalev, E.; Cervera, R.; Font, J.; Fridkin, M.; Thiesen, H.-J.; et al. The Efficacy of Specific IVIG Anti-Idiotypic Antibodies in Antiphospholipid Syndrome (APS): Trophoblast Invasiveness and APS Animal Model. Int. Immunol. 2007, 19, 857–865. [Google Scholar] [CrossRef]
  67. Jovanović, M.; Bozić, M.; Kovacević, T.; Radojcić, L.; Petronijević, M.; Vićovac, L. Effects of Anti-Phospholipid Antibodies on a Human Trophoblast Cell Line (HTR-8/SVneo). Acta Histochem. 2010, 112, 34–41. [Google Scholar] [CrossRef]
  68. Kovačević, T.M.; Radojčić, L.; Tošić, N.M.; Pavlović, S.T.; Vićovac, L.M. Monoclonal Antibody 26 Cross-Reactive with Β2-Glycoprotein I Affects Human Trophoblast Invasion in Vitro. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 171, 23–29. [Google Scholar] [CrossRef]
  69. Liu, J.; Zhang, L.; Tian, Y.; Wan, S.; Hu, M.; Song, S.; Zhang, M.; Zhou, Q.; Xia, Y.; Wang, X. Protection by Hydroxychloroquine Prevents Placental Injury in Obstetric Antiphospholipid Syndrome. J. Cell. Mol. Med. 2022, 26, 4357–4370. [Google Scholar] [CrossRef]
  70. Huppertz, B.; Kaufmann, P.; Kingdom, J. Trophoblast Turnover in Health and Disease. Fetal Matern. Med. Rev. 2002, 13, 103–118. [Google Scholar] [CrossRef]
  71. Sharp, A.N.; Heazell, A.E.P.; Crocker, I.P.; Mor, G. Placental Apoptosis in Health and Disease. Am. J. Reprod. Immunol. 2010, 64, 159–169. [Google Scholar] [CrossRef] [PubMed]
  72. Matalon, S.T.; Shoenfeld, Y.; Blank, M.; Yacobi, S.; Von Landenberg, P.; Ornoy, A. Antiphosphatidylserine Antibodies Affect Rat Yolk Sacs in Culture: A Mechanism for Fetal Loss in Antiphospholipid Syndrome. Am. J. Reprod. Immunol. 2004, 51, 144–151. [Google Scholar] [CrossRef] [PubMed]
  73. Ornoy, A.; Yacobi, S.; Matalon, S.T.; Blank, M.; Blumenfeld, Z.; Miller, R.K.; Shoenfeld, Y. The Effects of Antiphospholipid Antibodies Obtained from Women with SLE/APS and Associated Pregnancy Loss on Rat Embryos and Placental Explants in Culture. Lupus 2003, 12, 573–578. [Google Scholar] [CrossRef] [PubMed]
  74. Chu, H.; Sacharidou, A.; Nguyen, A.; Li, C.; Chambliss, K.L.; Salmon, J.E.; Shen, Y.-M.; Lo, J.; Leone, G.W.; Herz, J.; et al. Protein Phosphatase 2A Activation Via ApoER2 in Trophoblasts Drives Preeclampsia in a Mouse Model of the Antiphospholipid Syndrome. Circ. Res. 2021, 129, 735–750. [Google Scholar] [CrossRef] [PubMed]
  75. Velayuthaprabhu, S.; Matsubayashi, H.; Sugi, T.; Nakamura, M.; Ohnishi, Y.; Ogura, T.; Archunan, G. Expression of Apoptosis in Placenta of Experimental Antiphospholipid Syndrome Mouse. Am. J. Reprod. Immunol. 2013, 69, 486–494. [Google Scholar] [CrossRef] [PubMed]
  76. Bose, P.; Kadyrov, M.; Goldin, R.; Hahn, S.; Backos, M.; Regan, L.; Huppertz, B. Aberrations of Early Trophoblast Differentiation Predispose to Pregnancy Failure: Lessons from the Anti-Phospholipid Syndrome. Placenta 2006, 27, 869–875. [Google Scholar] [CrossRef]
  77. Yacobi, S.; Ornoy, A.; Blumenfeld, Z.; Miller, R.K. Effect of Sera from Women with Systemic Lupus Erythematosus or Antiphospholipid Syndrome and Recurrent Abortions on Human Placental Explants in Culture. Teratology 2002, 66, 300–308. [Google Scholar] [CrossRef]
  78. Chamley, L.W.; Holland, O.J.; Chen, Q.; Viall, C.A.; Stone, P.R.; Abumaree, M. Review: Where Is the Maternofetal Interface? Placenta 2014, 35, S74–S80. [Google Scholar] [CrossRef]
  79. Chen, Q.; Viall, C.; Kang, Y.; Liu, B.; Stone, P.; Chamley, L. Anti-Phospholipid Antibodies Increase Non-Apoptotic Trophoblast Shedding: A Contribution to the Pathogenesis of Pre-Eclampsia in Affected Women? Placenta 2009, 30, 767–773. [Google Scholar] [CrossRef]
  80. Pantham, P.; Heazell, A.E.P.; Mullard, G.; Begley, P.; Chen, Q.; Brown, M.; Dunn, W.B.; Chamley, L.W. Antiphospholipid Antibodies Alter Cell-Death-Regulating Lipid Metabolites in First and Third Trimester Human Placentae. Am. J. Reprod. Immunol. 2015, 74, 181–199. [Google Scholar] [CrossRef]
  81. Pantham, P.; Rosario, R.; Chen, Q.; Print, C.G.; Chamley, L.W. Transcriptomic Analysis of Placenta Affected by Antiphospholipid Antibodies: Following the TRAIL of Trophoblast Death. J. Reprod. Immunol. 2012, 94, 151–154. [Google Scholar] [CrossRef] [PubMed]
  82. di Simone, N.; Castellani, R.; Raschi, E.; Borghi, M.O.; Meroni, P.L.; Caruso, A. Anti-Beta-2 Glycoprotein I Antibodies Affect Bcl-2 and Bax Trophoblast Expression without Evidence of Apoptosis. Ann. N. Y. Acad. Sci. 2006, 1069, 364–376. [Google Scholar] [CrossRef] [PubMed]
  83. Jovanovic, M.; Kovacevic, T.; Stefanoska, I.; Vicovac, L. The Effect of IL-6 on the Trophoblast Cell Line HTR-8/SVneo. Arch. Biol. Sci. 2010, 62, 531–538. [Google Scholar] [CrossRef]
  84. Cohen, M.; Meisser, A.; Bischof, P. Metalloproteinases and Human Placental Invasiveness. Placenta 2006, 27, 783–793. [Google Scholar] [CrossRef] [PubMed]
  85. Damsky, C.H.; Fitzgerald, M.L.; Fisher, S.J. Distribution Patterns of Extracellular Matrix Components and Adhesion Receptors Are Intricately Modulated during First Trimester Cytotrophoblast Differentiation along the Invasive Pathway, in Vivo. J. Clin. Investig. 1992, 89, 210–222. [Google Scholar] [CrossRef]
  86. Zhou, Y.; Fisher, S.J.; Janatpour, M.; Genbacev, O.; Dejana, E.; Wheelock, M.; Damsky, C.H. Human Cytotrophoblasts Adopt a Vascular Phenotype as They Differentiate. A Strategy for Successful Endovascular Invasion? J. Clin. Investig. 1997, 99, 2139–2151. [Google Scholar] [CrossRef] [PubMed]
  87. Poulton, K.; Ripoll, V.M.; Pericleous, C.; Meroni, P.L.; Gerosa, M.; Ioannou, Y.; Rahman, A.; Giles, I.P. Purified IgG from Patients with Obstetric but Not IgG from Non-Obstetric Antiphospholipid Syndrome Inhibit Trophoblast Invasion. Am. J. Reprod. Immunol. 2015, 73, 390–401. [Google Scholar] [CrossRef]
  88. Di Simone, N.; Castellani, R.; Caliandro, D.; Caruso, A. Antiphospholid Antibodies Regulate the Expression of Trophoblast Cell Adhesion Molecules. Fertil. Steril. 2002, 77, 805–811. [Google Scholar] [CrossRef]
  89. Jovanović Krivokuća, M.; Abu Rabi, T.; Stefanoska, I.; Vrzić-Petronijević, S.; Petronijević, M.; Vićovac, L. Immunoglobulins from Sera of APS Patients Bind HTR-8/SVneo Trophoblast Cell Line and Reduce Additional Mediators of Cell Invasion. Reprod. Biol. 2017, 17, 389–395. [Google Scholar] [CrossRef]
  90. Cohen, M.; Bischof, P. Factors Regulating Trophoblast Invasion. Gynecol. Obstet. Investig. 2007, 64, 126–130. [Google Scholar] [CrossRef]
  91. Xu, P.; Alfaidy, N.; Challis, J.R.G. Expression of Matrix Metalloproteinase (MMP)-2 and MMP-9 in Human Placenta and Fetal Membranes in Relation to Preterm and Term Labor. J. Clin. Endocrinol. Metab. 2002, 87, 1353–1361. [Google Scholar] [CrossRef]
  92. Mulla, M.J.; Salmon, J.E.; Chamley, L.W.; Brosens, J.J.; Boeras, C.M.; Kavathas, P.B.; Abrahams, V.M. A Role for Uric Acid and the Nalp3 Inflammasome in Antiphospholipid Antibody-Induced IL-1β Production by Human First Trimester Trophoblast. PLoS ONE 2013, 8, e65237. [Google Scholar] [CrossRef] [PubMed]
  93. Gysler, S.M.; Mulla, M.J.; Guerra, M.; Brosens, J.J.; Salmon, J.E.; Chamley, L.W.; Abrahams, V.M. Antiphospholipid Antibody-Induced MiR-146a-3p Drives Trophoblast Interleukin-8 Secretion through Activation of Toll-like Receptor 8. Mol. Hum. Reprod. 2016, 22, 465–474. [Google Scholar] [CrossRef] [PubMed]
  94. Mulla, M.J.; Pasternak, M.C.; Salmon, J.E.; Chamley, L.W.; Abrahams, V.M. Role of NOD2 in Antiphospholipid Antibody-Induced and Bacterial MDP Amplification of Trophoblast Inflammation. J. Autoimmun. 2019, 98, 103–112. [Google Scholar] [CrossRef] [PubMed]
  95. Kolundžić, N.; Bojić-Trbojević, Ž.; Kovačević, T.; Stefanoska, I.; Kadoya, T.; Vićovac, L. Galectin-1 Is Part of Human Trophoblast Invasion Machinery—A Functional Study In Vitro. PLoS ONE 2011, 6, e28514. [Google Scholar] [CrossRef]
  96. Bojic-Trbojevic, Ž.; Jovanovic Krivokuca, M.; Stefanoska, I.; Kolundžic, N.; Vilotic, A.; Kadoya, T.; Vicovac, L. Integrin Β1 Is Bound to Galectin-1 in Human Trophoblast. J. Biochem. 2018, 163, 39–50. [Google Scholar] [CrossRef]
  97. LaMarca, H.L.; Dash, P.R.; Vishnuthevan, K.; Harvey, E.; Sullivan, D.E.; Morris, C.A.; Whitley, G.S.J. Epidermal Growth Factor-Stimulated Extravillous Cytotrophoblast Motility Is Mediated by the Activation of PI3-K, Akt and Both P38 and P42/44 Mitogen-Activated Protein Kinases. Hum. Reprod. 2008, 23, 1733–1741. [Google Scholar] [CrossRef]
  98. Alvarez, A.M.; Mulla, M.J.; Chamley, L.W.; Cadavid, A.P.; Abrahams, V.M. Aspirin-Triggered Lipoxin Prevents Antiphospholipid Antibody Effects on Human Trophoblast Migration and Endothelial Cell Interactions. Arthritis Rheumatol. 2015, 67, 488–497. [Google Scholar] [CrossRef]
  99. Bulla, R.; Villa, A.; Bossi, F.; Cassetti, A.; Radillo, O.; Spessotto, P.; De Seta, F.; Guaschino, S.; Tedesco, F. VE-Cadherin Is a Critical Molecule for Trophoblast-Endothelial Cell Interaction in Decidual Spiral Arteries. Exp. Cell Res. 2005, 303, 101–113. [Google Scholar] [CrossRef]
  100. Sung, D.C.; Chen, X.; Chen, M.; Yang, J.; Schultz, S.; Babu, A.; Xu, Y.; Gao, S.; Keller, T.C.S.; Mericko-Ishizuka, P.; et al. VE-Cadherin Enables Trophoblast Endovascular Invasion and Spiral Artery Remodeling during Placental Development. eLife 2022, 11, e77241. [Google Scholar] [CrossRef]
  101. Huang, Z.; Tang, Z.; Guan, H.; Leung, W.; Wang, L.; Xia, H.; Zhang, W. Inactivation of Yes-Associated Protein Mediates Trophoblast Dysfunction: A New Mechanism of Pregnancy Loss Associated with Anti-Phospholipid Antibodies? Biomedicines 2022, 10, 3296. [Google Scholar] [CrossRef] [PubMed]
  102. Sun, M.; Na, Q.; Huang, L.; Song, G.; Jin, F.; Li, Y.; Hou, Y.; Kang, D.; Qiao, C. YAP Is Decreased in Preeclampsia and Regulates Invasion and Apoptosis of HTR-8/SVneo. Reprod. Sci. 2018, 25, 1382–1393. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, R.; Wei, C.; Ma, Q.; Wang, W. Hippo-YAP1 Signaling Pathway and Severe Preeclampsia (SPE) in the Chinese Population. Pregnancy Hypertens. 2020, 19, 1–10. [Google Scholar] [CrossRef]
  104. Yue, C.; Chen, A.C.H.; Tian, S.; Fong, S.W.; Lee, K.C.; Zhang, J.; Ng, E.H.Y.; Lee, K.F.; Yeung, W.S.B.; Lee, Y.L. Human Embryonic Stem Cell-Derived Blastocyst-like Spheroids Resemble Human Trophectoderm during Early Implantation Process. Fertil. Steril. 2020, 114, 653–664.e6. [Google Scholar] [CrossRef] [PubMed]
  105. Adler, R.R.; Ng, A.K.; Rote, N.S. Monoclonal Antiphosphatidylserine Antibody Inhibits Intercellular Fusion of the Choriocarcinoma Line, JAR. Biol. Reprod. 1995, 53, 905–910. [Google Scholar] [CrossRef] [PubMed]
  106. Marchetti, T.; Ruffatti, A.; Wuillemin, C.; de Moerloose, P.; Cohen, M. Hydroxychloroquine Restores Trophoblast Fusion Affected by Antiphospholipid Antibodies. J. Thromb. Haemost. 2014, 12, 910–920. [Google Scholar] [CrossRef] [PubMed]
  107. Shurtz-Swirski, R.; Inbar, O.; Blank, M.; Cohen, J.; Bakimer, R.; Barnea, E.R.; Shoenfeld, Y. In Vitro Effect of Anticardiolipin Autoantibodies upon Total and Pulsatile Placental HCG Secretion during Early Pregnancy. Am. J. Reprod. Immunol. 1993, 29, 206–210. [Google Scholar] [CrossRef] [PubMed]
  108. Zussman, R.; Xu, L.Y.; Damani, T.; Groom, K.M.; Chen, Q.; Seers, B.; Viall, C.A.; Chamley, L.W.; Hickey, A. Antiphospholipid Antibodies Can Specifically Target Placental Mitochondria and Induce ROS Production. J. Autoimmun. 2020, 111, 102437. [Google Scholar] [CrossRef]
  109. Alarcón-Segovia, D.; Ruíz-Argüelles, A.; Fishbein, E. Antibody Penetration into Living Cells. I. Intranuclear Immunoglobulin in Peripheral Blood Mononuclear Cells in Mixed Connective Tissue Disease and Systemic Lupus Erythematosus. Clin. Exp. Immunol. 1979, 35, 364–375. [Google Scholar]
  110. Galve-de Rochemonteix, B.; Kobayashi, T.; Rosnoblet, C.; Lindsay, M.; Parton, R.G.; Reber, G.; de Maistre, E.; Wahl, D.; Kruithof, E.K.; Gruenberg, J.; et al. Interaction of Anti-Phospholipid Antibodies with Late Endosomes of Human Endothelial Cells. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 563–574. [Google Scholar] [CrossRef]
  111. Hou, S.; Fölsch, H.; Ke, K.; Cook Mills, J.; Ramsey-Goldman, R.; Zhao, M. Early Endosome as a Pathogenic Target for Antiphosphatidylethanolamine Antibodies. Proc. Natl. Acad. Sci. USA 2017, 114, 13798–13803. [Google Scholar] [CrossRef] [PubMed]
  112. Virachith, S.; Saito, M.; Watanabe, Y.; Inoue, K.; Hoshi, O.; Kubota, T. Anti-Β2-Glycoprotein I Antibody with DNA Binding Activity Enters Living Monocytes via Cell Surface DNA and Induces Tissue Factor Expression. Clin. Exp. Immunol. 2019, 195, 167–178. [Google Scholar] [CrossRef] [PubMed]
  113. Jovanović Krivokuća, M.; Stefanoska, I.; Abu Rabi, T.; Marković, M.; Janković, S.; Vrzić-Petronijević, S.; Vićovac, L. Immunoglobulins from Sera of Antiphospholipid Syndrome Patients Are Internalized in the HTR-8/SVneo Cell Line and Cytotrophoblast in Culture. Immunobiology 2018, 223, 544–548. [Google Scholar] [CrossRef] [PubMed]
  114. Viall, C.A.; Chen, Q.; Stone, P.R.; Chamley, L.W. Human Extravillous Trophoblasts Bind but Do Not Internalize Antiphospholipid Antibodies. Placenta 2016, 42, 9–16. [Google Scholar] [CrossRef]
  115. Pérez-Sánchez, C.; Arias-de la Rosa, I.; Aguirre, M.Á.; Luque-Tévar, M.; Ruiz-Limón, P.; Barbarroja, N.; Jiménez-Gómez, Y.; Ábalos-Aguilera, M.C.; Collantes-Estévez, E.; Segui, P.; et al. Circulating MicroRNAs as Biomarkers of Disease and Typification of the Atherothrombotic Status in Antiphospholipid Syndrome. Haematologica 2018, 103, 908–918. [Google Scholar] [CrossRef]
  116. Juárez-Vicuña, Y.; Guzmán-Martín, C.A.; Martínez-Martínez, L.A.; Hernández-Díazcouder, A.; Huesca-Gómez, C.; Gamboa, R.; Amezcua-Guerra, L.M.; Chacon-Perez, M.; Amigo, M.C.; Sánchez-Muñoz, F. MiR-19b-3p and MiR-20a-5p Are Associated with the Levels of Antiphospholipid Antibodies in Patients with Antiphospholipid Syndrome. Rheumatol. Int. 2021, 41, 1329–1335. [Google Scholar] [CrossRef]
  117. Pérez-Sánchez, L.; Patiño-Trives, A.M.; Aguirre-Zamorano, M.Á.; Luque-Tévar, M.; Ábalos-Aguilera, M.C.; Arias-de la Rosa, I.; Seguí, P.; Velasco-Gimena, F.; Barbarroja, N.; Escudero-Contreras, A.; et al. Characterization of Antiphospholipid Syndrome Atherothrombotic Risk by Unsupervised Integrated Transcriptomic Analyses. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 865–877. [Google Scholar] [CrossRef]
  118. Guzmán-Martín, C.A.; Juárez-Vicuña, Y.; Domínguez-López, A.; González-Ramírez, J.; Amezcua-Guerra, L.M.; Martínez-Martínez, L.A.; Sánchez-Muñoz, F. LncRNAs Dysregulation in Monocytes from Primary Antiphospholipid Syndrome Patients: A Bioinformatic and an Experimental Proof-of-Concept Approach. Mol. Biol. Rep. 2023, 50, 937–941. [Google Scholar] [CrossRef]
  119. Zou, Y.; Xu, H. Involvement of Long Noncoding RNAs in the Pathogenesis of Autoimmune Diseases. J. Transl. Autoimmun. 2020, 3, 100044. [Google Scholar] [CrossRef]
  120. Zhang, L.; Wu, H.; Zhao, M.; Chang, C.; Lu, Q. Clinical Significance of MiRNAs in Autoimmunity. J. Autoimmun. 2020, 109, 102438. [Google Scholar] [CrossRef]
  121. Wu, H.; Chen, S.; Li, A.; Shen, K.; Wang, S.; Wang, S.; Wu, P.; Luo, W.; Pan, Q. LncRNA Expression Profiles in Systemic Lupus Erythematosus and Rheumatoid Arthritis: Emerging Biomarkers and Therapeutic Targets. Front. Immunol. 2021, 12, 792884. [Google Scholar] [CrossRef] [PubMed]
  122. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  123. Zhang, X.; Wang, W.; Zhu, W.; Dong, J.; Cheng, Y.; Yin, Z.; Shen, F. Mechanisms and Functions of Long Non-Coding RNAs at Multiple Regulatory Levels. Int. J. Mol. Sci. 2019, 20, 5573. [Google Scholar] [CrossRef] [PubMed]
  124. Cavalcante, G.C.; Magalhães, L.; Ribeiro-Dos-Santos, Â.; Vidal, A.F. Mitochondrial Epigenetics: Non-Coding RNAs as a Novel Layer of Complexity. Int. J. Mol. Sci. 2020, 21, 1838. [Google Scholar] [CrossRef]
  125. Ghafouri-Fard, S.; Shoorei, H.; Anamag, F.T.; Taheri, M. The Role of Non-Coding RNAs in Controlling Cell Cycle Related Proteins in Cancer Cells. Front. Oncol. 2020, 10, 608975. [Google Scholar] [CrossRef]
  126. Jiang, N.; Zhang, X.; Gu, X.; Li, X.; Shang, L. Progress in Understanding the Role of LncRNA in Programmed Cell Death. Cell Death Discov. 2021, 7, 30. [Google Scholar] [CrossRef]
  127. Taganov, K.D.; Boldin, M.P.; Chang, K.-J.; Baltimore, D. NF-ΚB-Dependent Induction of MicroRNA MiR-146, an Inhibitor Targeted to Signaling Proteins of Innate Immune Responses. Proc. Natl. Acad. Sci. USA 2006, 103, 12481–12486. [Google Scholar] [CrossRef]
  128. Tang, B.; Xiao, B.; Liu, Z.; Li, N.; Zhu, E.-D.; Li, B.-S.; Xie, Q.-H.; Zhuang, Y.; Zou, Q.-M.; Mao, X.-H. Identification of MyD88 as a Novel Target of MiR-155, Involved in Negative Regulation of Helicobacter Pylori-Induced Inflammation. FEBS Lett. 2010, 584, 1481–1486. [Google Scholar] [CrossRef]
  129. Qi, J.; Qiao, Y.; Wang, P.; Li, S.; Zhao, W.; Gao, C. MicroRNA-210 Negatively Regulates LPS-Induced Production of Proinflammatory Cytokines by Targeting NF-ΚB1 in Murine Macrophages. FEBS Lett. 2012, 586, 1201–1207. [Google Scholar] [CrossRef]
  130. Arenas-Padilla, M.; Mata-Haro, V. Regulation of TLR Signaling Pathways by MicroRNAs: Implications in Inflammatory Diseases. Cent. J. Immunol. 2018, 43, 482–489. [Google Scholar] [CrossRef]
  131. Yang, C.; Lim, W.; Park, J.; Park, S.; You, S.; Song, G. Anti-Inflammatory Effects of Mesenchymal Stem Cell-Derived Exosomal MicroRNA-146a-5p and MicroRNA-548e-5p on Human Trophoblast Cells. Mol. Hum. Reprod. 2019, 25, 755–771. [Google Scholar] [CrossRef]
  132. Qi, Y.; Cui, S.; Liu, L.; Liu, B.; Wang, T.; Yan, S.; Tian, H.; Huang, X. Expression and Role of MiR-146a and SMAD4 in Placental Tissue of Pregnant Women with Preeclampsia. J. Obstet. Gynaecol. Res. 2022, 48, 2151–2161. [Google Scholar] [CrossRef] [PubMed]
  133. Xiao, C.; Rui, Y.; Zhou, S.; Huang, Y.; Wei, Y.; Wang, Z. TNF-Related Apoptosis-Inducing Ligand (TRAIL) Promotes Trophoblast Cell Invasion via MiR-146a-EGFR/CXCR4 Axis: A Novel Mechanism for Preeclampsia? Placenta 2020, 93, 8–16. [Google Scholar] [CrossRef] [PubMed]
  134. Ding, J.; Zhang, Y.; Cai, X.; Zhang, Y.; Yan, S.; Wang, J.; Zhang, S.; Yin, T.; Yang, C.; Yang, J. Extracellular Vesicles Derived from M1 Macrophages Deliver MiR-146a-5p and MiR-146b-5p to Suppress Trophoblast Migration and Invasion by Targeting TRAF6 in Recurrent Spontaneous Abortion. Theranostics 2021, 11, 5813–5830. [Google Scholar] [CrossRef]
  135. Peng, P.; Song, H.; Xie, C.; Zheng, W.; Ma, H.; Xin, D.; Zhan, J.; Yuan, X.; Chen, A.; Tao, J.; et al. MiR-146a-5p-Mediated Suppression on Trophoblast Cell Progression and Epithelial-Mesenchymal Transition in Preeclampsia. Biol. Res. 2021, 54, 30. [Google Scholar] [CrossRef] [PubMed]
  136. Yu, Y.; An, X.; Fan, D. Histone Deacetylase Sirtuin 2 Enhances Viability of Trophoblasts Through P65-Mediated MicroRNA-146a/ACKR2 Axis. Reprod. Sci. 2021, 28, 1370–1381. [Google Scholar] [CrossRef]
  137. Zhang, Y.; Diao, Z.; Su, L.; Sun, H.; Li, R.; Cui, H.; Hu, Y. MicroRNA-155 Contributes to Preeclampsia by down-Regulating CYR61. Am. J. Obstet. Gynecol. 2010, 202, 466.e1–466.e7. [Google Scholar] [CrossRef]
  138. Dai, Y.; Diao, Z.; Sun, H.; Li, R.; Qiu, Z.; Hu, Y. MicroRNA-155 Is Involved in the Remodelling of Human-Trophoblast-Derived HTR-8/SVneo Cells Induced by Lipopolysaccharides. Hum. Reprod. 2011, 26, 1882–1891. [Google Scholar] [CrossRef]
  139. Li, X.; Li, C.; Dong, X.; Gou, W. MicroRNA-155 Inhibits Migration of Trophoblast Cells and Contributes to the Pathogenesis of Severe Preeclampsia by Regulating Endothelial Nitric Oxide Synthase. Mol. Med. Rep. 2014, 10, 550–554. [Google Scholar] [CrossRef]
  140. Anton, L.; Olarerin-George, A.O.; Schwartz, N.; Srinivas, S.; Bastek, J.; Hogenesch, J.B.; Elovitz, M.A. MiR-210 Inhibits Trophoblast Invasion and Is a Serum Biomarker for Preeclampsia. Am. J. Pathol. 2013, 183, 1437–1445. [Google Scholar] [CrossRef]
  141. Li, L.; Huang, X.; He, Z.; Xiong, Y.; Fang, Q. MiRNA-210-3p Regulates Trophoblast Proliferation and Invasiveness through Fibroblast Growth Factor 1 in Selective Intrauterine Growth Restriction. J. Cell. Mol. Med. 2019, 23, 4422–4433. [Google Scholar] [CrossRef] [PubMed]
  142. Hayder, H.; Fu, G.; Nadeem, L.; O’Brien, J.A.; Lye, S.J.; Peng, C. Overexpression of MiR-210-3p Impairs Extravillous Trophoblast Functions Associated with Uterine Spiral Artery Remodeling. Int. J. Mol. Sci. 2021, 22, 3961. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, R.; Zou, L.; Yang, X. MicroRNA-210/ Long Non-Coding RNA MEG3 Axis Inhibits Trophoblast Cell Migration and Invasion by Suppressing EMT Process. Placenta 2021, 109, 64–71. [Google Scholar] [CrossRef]
  144. Testa, U.; Pelosi, E.; Castelli, G.; Labbaye, C. MiR-146 and MiR-155: Two Key Modulators of Immune Response and Tumor Development. Non-Coding RNA 2017, 3, 22. [Google Scholar] [CrossRef]
  145. Kobayashi, T.; Walsh, M.C.; Choi, Y. The Role of TRAF6 in Signal Transduction and the Immune Response. Microbes Infect. 2004, 6, 1333–1338. [Google Scholar] [CrossRef]
  146. Wang, J.; Wu, X.; Jiang, M.; Tai, G. Mechanism by Which TRAF6 Participates in the Immune Regulation of Autoimmune Diseases and Cancer. Biomed Res. Int. 2020, 2020, 4607197. [Google Scholar] [CrossRef]
  147. Gowhari Shabgah, A.; Jadidi-Niaragh, F.; Mohammadi, H.; Ebrahimzadeh, F.; Oveisee, M.; Jahanara, A.; Gholizadeh Navashenaq, J. The Role of Atypical Chemokine Receptor D6 (ACKR2) in Physiological and Pathological Conditions; Friend, Foe, or Both? Front. Immunol. 2022, 13, 861931. [Google Scholar] [CrossRef] [PubMed]
  148. Martinez de la Torre, Y.; Buracchi, C.; Borroni, E.M.; Dupor, J.; Bonecchi, R.; Nebuloni, M.; Pasqualini, F.; Doni, A.; Lauri, E.; Agostinis, C.; et al. Protection against Inflammation- and Autoantibody-Caused Fetal Loss by the Chemokine Decoy Receptor D6. Proc. Natl. Acad. Sci. USA 2007, 104, 2319–2324. [Google Scholar] [CrossRef]
  149. Yan, S.; Cui, S.; Zhang, L.; Yang, B.; Yuan, Y.; Lv, X.; Fu, H.; Li, Y.; Huang, C.; Wang, P. Expression of ACKR2 in Placentas from Different Types of Preeclampsia. Placenta 2020, 90, 121–127. [Google Scholar] [CrossRef]
  150. Knöfler, M. Critical Growth Factors and Signalling Pathways Controlling Human Trophoblast Invasion. Int. J. Dev. Biol. 2010, 54, 269–280. [Google Scholar] [CrossRef]
  151. Wang, L.; Li, X.; Zhao, Y.; Fang, C.; Lian, Y.; Gou, W.; Han, T.; Zhu, X. Insights into the Mechanism of CXCL12-Mediated Signaling in Trophoblast Functions and Placental Angiogenesis. Acta Biochim. Biophys. Sin. 2015, 47, 663–672. [Google Scholar] [CrossRef] [PubMed]
  152. Li, Y.; Yan, J.; Chang, H.-M.; Chen, Z.-J.; Leung, P.C.K. Roles of TGF-β Superfamily Proteins in Extravillous Trophoblast Invasion. Trends Endocrinol. Metab. 2021, 32, 170–189. [Google Scholar] [CrossRef] [PubMed]
  153. Wu, J.-X.; Shi, M.; Gong, B.-M.; Ji, B.-W.; Hu, C.-C.; Wang, G.-C.; Lei, L.; Tang, C.; Sun, L.V.; Wu, X.-H.; et al. An MiRNA-MRNA Integrative Analysis in Human Placentas and Mice: Role of the Smad2/MiR-155-5p Axis in the Development of Fetal Growth Restriction. Front. Bioeng. Biotechnol. 2023, 11, 1159805. [Google Scholar] [CrossRef] [PubMed]
  154. Dai, Y.; Qiu, Z.; Diao, Z.; Shen, L.; Xue, P.; Sun, H.; Hu, Y. MicroRNA-155 Inhibits Proliferation and Migration of Human Extravillous Trophoblast Derived HTR-8/SVneo Cells via down-Regulating Cyclin D1. Placenta 2012, 33, 824–829. [Google Scholar] [CrossRef]
  155. Luo, X.; Pan, C.; Guo, X.; Gu, C.; Huang, Y.; Guo, J.; Zeng, Y.; Yue, J.; Cui, S. Methylation Mediated Silencing of MiR-155 Suppresses the Development of Preeclampsia In Vitro and In Vivo by Targeting FOXO3. Mediat. Inflamm. 2022, 2022, 4250621. [Google Scholar] [CrossRef]
  156. Chan, Y.C.; Banerjee, J.; Choi, S.Y.; Sen, C.K. MiR-210: The Master Hypoxamir. Microcirculation 2012, 19, 215–223. [Google Scholar] [CrossRef]
  157. Anton, L.; DeVine, A.; Polyak, E.; Olarerin-George, A.; Brown, A.G.; Falk, M.J.; Elovitz, M.A. HIF-1α Stabilization Increases MiR-210 Eliciting First Trimester Extravillous Trophoblast Mitochondrial Dysfunction. Front. Physiol. 2019, 10, 699. [Google Scholar] [CrossRef]
  158. Wang, Y.; Liu, H.-Z.; Liu, Y.; Wang, H.-J.; Pang, W.-W.; Zhang, J.-J. Downregulated MALAT1 Relates to Recurrent Pregnancy Loss via Sponging MiRNAs. Kaohsiung J. Med. Sci. 2018, 34, 503–510. [Google Scholar] [CrossRef]
  159. Bahia, W.; Soltani, I.; Abidi, A.; Haddad, A.; Ferchichi, S.; Menif, S.; Almawi, W.Y. Identification of Genes and MiRNA Associated with Idiopathic Recurrent Pregnancy Loss: An Exploratory Data Mining Study. BMC Med. Genom. 2020, 13, 75. [Google Scholar] [CrossRef]
  160. Huang, Y.; Hao, J.; Liao, Y.; Zhou, L.; Wang, K.; Zou, H.; Hu, Y.; Li, J. Transcriptome Sequencing Identified the CeRNA Network Associated with Recurrent Spontaneous Abortion. BMC Med. Genom. 2021, 14, 278. [Google Scholar] [CrossRef]
  161. Pineles, B.L.; Romero, R.; Montenegro, D.; Tarca, A.L.; Han, Y.M.; Kim, Y.M.; Draghici, S.; Espinoza, J.; Kusanovic, J.P.; Mittal, P.; et al. Distinct Subsets of MicroRNAs Are Expressed Differentially in the Human Placentas of Patients with Preeclampsia. Am. J. Obstet. Gynecol. 2007, 196, 261.e1–261.e6. [Google Scholar] [CrossRef] [PubMed]
  162. Enquobahrie, D.A.; Abetew, D.F.; Sorensen, T.K.; Willoughby, D.; Chidambaram, K.; Williams, M.A. Placental MicroRNA Expression in Pregnancies Complicated by Preeclampsia. Am. J. Obstet. Gynecol. 2011, 204, 178.e12–178.e21. [Google Scholar] [CrossRef]
  163. Muralimanoharan, S.; Maloyan, A.; Mele, J.; Guo, C.; Myatt, L.G.; Myatt, L. MIR-210 Modulates Mitochondrial Respiration in Placenta with Preeclampsia. Placenta 2012, 33, 816–823. [Google Scholar] [CrossRef] [PubMed]
  164. Hromadnikova, I.; Kotlabova, K.; Krofta, L. Cardiovascular Disease-Associated MicroRNA Dysregulation during the First Trimester of Gestation in Women with Chronic Hypertension and Normotensive Women Subsequently Developing Gestational Hypertension or Preeclampsia with or without Fetal Growth Restriction. Biomedicines 2022, 10, 256. [Google Scholar] [CrossRef]
  165. Hromadnikova, I.; Kotlabova, K.; Krofta, L. First-Trimester Screening for Miscarriage or Stillbirth-Prediction Model Based on MicroRNA Biomarkers. Int. J. Mol. Sci. 2023, 24, 10137. [Google Scholar] [CrossRef]
  166. Kim, S.; Park, M.; Kim, J.-Y.; Kim, T.; Hwang, J.Y.; Ha, K.-S.; Won, M.-H.; Ryoo, S.; Kwon, Y.-G.; Kim, Y.-M. Circulating MiRNAs Associated with Dysregulated Vascular and Trophoblast Function as Target-Based Diagnostic Biomarkers for Preeclampsia. Cells 2020, 9, 2003. [Google Scholar] [CrossRef]
  167. Gan, L.; Liu, Z.; Wei, M.; Chen, Y.; Yang, X.; Chen, L.; Xiao, X. MiR-210 and MiR-155 as Potential Diagnostic Markers for Pre-Eclampsia Pregnancies. Medicine 2017, 96, e7515. [Google Scholar] [CrossRef]
  168. Rai, R.; Regan, L. Recurrent Miscarriage. Lancet 2006, 368, 601–611. [Google Scholar] [CrossRef]
  169. Wu, H.-Y.; Wang, X.-H.; Liu, K.; Zhang, J.-L. LncRNA MALAT1 Regulates Trophoblast Cells Migration and Invasion via MiR-206/IGF-1 Axis. Cell Cycle 2020, 19, 39–52. [Google Scholar] [CrossRef]
  170. Li, Q.; Wang, T.; Huang, S.; Zuo, Q.; Jiang, Z.; Yang, N.; Sun, L. LncRNA MALAT1 Affects the Migration and Invasion of Trophoblast Cells by Regulating FOS Expression in Early-Onset Preeclampsia. Pregnancy Hypertens. 2020, 21, 50–57. [Google Scholar] [CrossRef]
  171. Feng, C.; Cheng, L.; Jin, J.; Liu, X.; Wang, F. Long Non-Coding RNA MALAT1 Regulates Trophoblast Functions through VEGF/VEGFR1 Signaling Pathway. Arch. Gynecol. Obstet. 2021, 304, 873–882. [Google Scholar] [CrossRef]
  172. Han, Y.; Wang, Y.; Zhang, C.; Li, Y.; Guo, J.; Tian, C. Metastasis-Associated Lung Adenocarcinoma Transcript 1 Induces Methyl-CpG-Binding Domain Protein 4 in Mice with Recurrent Spontaneous Abortion Caused by Anti-Phospholipid Antibody Positivity. Placenta 2023, 137, 38–48. [Google Scholar] [CrossRef]
  173. Li, Q.; Li, S.; Ding, J.; Pang, B.; Li, R.; Cao, H.; Ling, L. MALAT1 Modulates Trophoblast Phenotype via MiR-101-3p/VEGFA Axis. Arch. Biochem. Biophys. 2023, 744, 109692. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, Y.; Liu, H.-Z.; Liu, Y.; Wang, H.-J.; Pang, W.-W.; Zhang, J.-J. Disordered P53-MALAT1 Pathway Is Associated with Recurrent Miscarriage. Kaohsiung J. Med. Sci. 2019, 35, 87–94. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, H.; Meng, T.; Liu, X.; Sun, M.; Tong, C.; Liu, J.; Wang, H.; Du, J. Long Non-Coding RNA MALAT-1 Is Downregulated in Preeclampsia and Regulates Proliferation, Apoptosis, Migration and Invasion of JEG-3 Trophoblast Cells. Int. J. Clin. Exp. Pathol. 2015, 8, 12718–12727. [Google Scholar] [PubMed]
  176. Ala, U. Competing Endogenous RNAs, Non-Coding RNAs and Diseases: An Intertwined Story. Cells 2020, 9, 1574. [Google Scholar] [CrossRef]
  177. Ding, Y.; Guo, F.; Zhu, T.; Li, J.; Gu, D.; Jiang, W.; Lu, Y.; Zhou, D. Mechanism of Long Non-Coding RNA MALAT1 in Lipopolysaccharide-Induced Acute Kidney Injury Is Mediated by the MiR-146a/NF-ΚB Signaling Pathway. Int. J. Mol. Med. 2018, 41, 446–454. [Google Scholar] [CrossRef]
  178. Peng, N.; He, J.; Li, J.; Huang, H.; Huang, W.; Liao, Y.; Zhu, S. Long Noncoding RNA MALAT1 Inhibits the Apoptosis and Autophagy of Hepatocellular Carcinoma Cell by Targeting the MicroRNA-146a/PI3K/Akt/MTOR Axis. Cancer Cell Int. 2020, 20, 165. [Google Scholar] [CrossRef]
  179. Sheng, X.-F.; Hong, L.-L.; Li, H.; Huang, F.-Y.; Wen, Q.; Zhuang, H.-F. Long Non-Coding RNA MALAT1 Modulate Cell Migration, Proliferation and Apoptosis by Sponging MicroRNA-146a to Regulate CXCR4 Expression in Acute Myeloid Leukemia. Hematology 2021, 26, 43–52. [Google Scholar] [CrossRef]
  180. Pei, C.; Gong, X.; Zhang, Y. LncRNA MALAT-1 Promotes Growth and Metastasis of Epithelial Ovarian Cancer via Sponging Microrna-22. Am. J. Transl. Res. 2020, 12, 6977–6987. [Google Scholar] [PubMed]
  181. Shi, C.; Ren, S.; Zhao, X.; Li, Q. LncRNA MALAT1 Regulates the Resistance of Breast Cancer Cells to Paclitaxel via the MiR-497-5p/SHOC2 Axis. Pharmacogenomics 2022, 23, 973–985. [Google Scholar] [CrossRef] [PubMed]
  182. Hajibabaei, S.; Nafissi, N.; Azimi, Y.; Mahdian, R.; Rahimi-Jamnani, F.; Valizadeh, V.; Rafiee, M.H.; Azizi, M. Targeting Long Non-Coding RNA MALAT1 Reverses Cancerous Phenotypes of Breast Cancer Cells through MicroRNA-561-3p/TOP2A Axis. Sci. Rep. 2023, 13, 8652. [Google Scholar] [CrossRef] [PubMed]
  183. Cao, S.; Wang, Y.; Li, J.; Lv, M.; Niu, H.; Tian, Y. Tumor-Suppressive Function of Long Noncoding RNA MALAT1 in Glioma Cells by Suppressing MiR-155 Expression and Activating FBXW7 Function. Am. J. Cancer Res. 2016, 6, 2561–2574. [Google Scholar] [PubMed]
  184. Li, S.; Sun, Y.; Zhong, L.; Xiao, Z.; Yang, M.; Chen, M.; Wang, C.; Xie, X.; Chen, X. The Suppression of Ox-LDL-Induced Inflammatory Cytokine Release and Apoptosis of HCAECs by Long Non-Coding RNA-MALAT1 via Regulating MicroRNA-155/SOCS1 Pathway. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 1175–1187. [Google Scholar] [CrossRef] [PubMed]
  185. Zhou, Q.; Lian, Y.; Zhang, Y.; Li, L.; Li, H.; Shen, D.; Zhou, Y.; Zhang, M.; Lu, Y.; Liu, J.; et al. Platelet-Derived Microparticles from Recurrent Miscarriage Associated with Antiphospholipid Antibody Syndrome Influence Behaviours of Trophoblast and Endothelial Cells. Mol. Hum. Reprod. 2019, 25, 483–494. [Google Scholar] [CrossRef] [PubMed]
  186. Li, X.; Wang, Q. Platelet-Derived Microparticles and Autoimmune Diseases. Int. J. Mol. Sci. 2023, 24, 10275. [Google Scholar] [CrossRef]
  187. Lugo-Gavidia, L.M.; Burger, D.; Matthews, V.B.; Nolde, J.M.; Galindo Kiuchi, M.; Carnagarin, R.; Kannenkeril, D.; Chan, J.; Joyson, A.; Herat, L.Y.; et al. Role of Microparticles in Cardiovascular Disease: Implications for Endothelial Dysfunction, Thrombosis, and Inflammation. Hypertension 2021, 77, 1825–1844. [Google Scholar] [CrossRef]
  188. Pan, Y.; Wang, Y.; Wang, Y.; Xu, S.; Jiang, F.; Han, Y.; Hu, M.; Liu, Z. Platelet-Derived Microvesicles (PMVs) in Cancer Progression and Clinical Applications. Clin. Transl. Oncol. 2023, 25, 873–881. [Google Scholar] [CrossRef]
  189. Laffont, B.; Corduan, A.; Plé, H.; Duchez, A.-C.; Cloutier, N.; Boilard, E.; Provost, P. Activated Platelets Can Deliver MRNA Regulatory Ago2•microRNA Complexes to Endothelial Cells via Microparticles. Blood 2013, 122, 253–261. [Google Scholar] [CrossRef]
  190. Provost, P. The Clinical Significance of Platelet Microparticle-Associated MicroRNAs. Clin. Chem. Lab. Med. 2017, 55, 657–666. [Google Scholar] [CrossRef]
  191. Wang, D.; Tian, Y.; Wang, S.; Li, Y.; Li, H.; Jiang, N.; Xie, Y.; Yu, M.; Li, A.; Wang, X.; et al. Antisense Oligonuleotides Influences Trophoblasts Behaviors by Changing LncNR_040117 Expression in Antiphospholipid Antibody Syndrome-Induced Recurrent Pregnancy Loss. J. Matern. Fetal. Neonatal Med. 2023, 36, 2183083. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A). Schematic presentation of the anchoring chorionic villus and trophoblast invasion into the decidual tissue in healthy pregnancy. (B). Possible mechanisms of antiphospholipid antibodies’ action on trophoblast cells leading to defective placentation/placental malfunction. aPL—antiphospholipid antibodies; CT—cytotrophoblast; ST—syncytiotrophoblast; EVT—extravillous trophoblast; SA—spiral artery; MMP-matrix metalloproteinase; Gal-1—galectin-1; βhCG—β-human chorionic gonadotropin.
Figure 1. (A). Schematic presentation of the anchoring chorionic villus and trophoblast invasion into the decidual tissue in healthy pregnancy. (B). Possible mechanisms of antiphospholipid antibodies’ action on trophoblast cells leading to defective placentation/placental malfunction. aPL—antiphospholipid antibodies; CT—cytotrophoblast; ST—syncytiotrophoblast; EVT—extravillous trophoblast; SA—spiral artery; MMP-matrix metalloproteinase; Gal-1—galectin-1; βhCG—β-human chorionic gonadotropin.
Biomedicines 11 02681 g001
Table 1. Criteria for diagnosis of APS.
Table 1. Criteria for diagnosis of APS.
APS Disease Classification Criteria According to Sydney Protocol
Clinical Criteria (at Least 1 of 2)Laboratory Criteria (at Least 1 of 3)
Vascular thrombosis:
  • ≥1 clinical episode of thrombosis in any tissue/organ, arterial or venous

Pregnancy morbidity:
  • ≥1 morphologically normal fetal loss, ≥10th week of gestation, or
  • ≥1 premature birth of a normal neonate before the 34th week due to (i) eclampsia or severe preeclampsia or (ii) placental insufficiency, or
  • ≥3 unexplained consecutive spontaneous abortions < 10th week of gestation (with exclusion of parental anatomic, hormonal or chromosomal causes).
Presence of (at least twice in min. 12 weeks):
  • Lupus coagulant (LA)
  • Antibody to cardiolipin (aCL), β-2 glycoprotein I (anti-β2GPI) (high titer, IgG or IgM)

Classification based on laboratory tests:
  • Type I: >1 laboratory criterion present (any combination)
  • Type IIa: LA antibodies only
  • Type IIb: aCL antibodies only
  • Type IIc: anti-β2GPI antibody only
Table 2. Possible implication of APS-associated ncRNAs in trophoblast cell function.
Table 2. Possible implication of APS-associated ncRNAs in trophoblast cell function.
ncRNAAPS-Associated ncRNA
Dysregulation
Implication in Trophoblast Function
miR-146a-5p
miR-146a-3p
miR-155
miR-210
Upregulated in anti-β2GPI treated HTR-8/SVneo cells [93]Overexpression of miR-146a [133,134,135,136], miR-155 [137,138,139] and miR-210 [140,141,142,143] inhibited invasion and migration of HTR-8/SVneo cells
Overexpression of miR-210 inhibited
mitochondrial respiration in primary EVT cells [157]
Overexpression of miR-210-3pinhibited tube formation of HTR-8/SVneo cells [142]
lncRNA MALAT1Downregulated in placentas of
APS-induced RPL patients [172]
MALAT1 downregulation inhibited proliferation, migration and invasion of HTR-8/SVneo cells [158,169,170,171,172]
LncNR_040117Upregulated in PMPs isolated from APS-induced RPL patients [24]LncNR_040117 downregulation stimulated proliferation, migration and invasion of HTR-8/SVneo cells [185,191]
EVT—extravillous trophoblast; PMPs—platelet-derived microparticles; RPL—recurrent pregnancy loss.
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

Vrzić Petronijević, S.; Vilotić, A.; Bojić-Trbojević, Ž.; Kostić, S.; Petronijević, M.; Vićovac, L.; Jovanović Krivokuća, M. Trophoblast Cell Function in the Antiphospholipid Syndrome. Biomedicines 2023, 11, 2681. https://doi.org/10.3390/biomedicines11102681

AMA Style

Vrzić Petronijević S, Vilotić A, Bojić-Trbojević Ž, Kostić S, Petronijević M, Vićovac L, Jovanović Krivokuća M. Trophoblast Cell Function in the Antiphospholipid Syndrome. Biomedicines. 2023; 11(10):2681. https://doi.org/10.3390/biomedicines11102681

Chicago/Turabian Style

Vrzić Petronijević, Svetlana, Aleksandra Vilotić, Žanka Bojić-Trbojević, Sanja Kostić, Miloš Petronijević, Ljiljana Vićovac, and Milica Jovanović Krivokuća. 2023. "Trophoblast Cell Function in the Antiphospholipid Syndrome" Biomedicines 11, no. 10: 2681. https://doi.org/10.3390/biomedicines11102681

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