Open Access

Importance of STAT3 signaling in preeclampsia (Review)

  • Authors:
    • Daniela Marzioni
    • Federica Piani
    • Nicoletta Di Simone
    • Stefano Raffaele Giannubilo
    • Andrea Ciavattini
    • Giovanni Tossetta
  • View Affiliations

  • Published online on: February 6, 2025     https://doi.org/10.3892/ijmm.2025.5499
  • Article Number: 58
  • Copyright : © Marzioni et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Placentation is a key process that is tightly regulated that ensures the normal placenta and fetal development. Preeclampsia (PE) is a hypertensive pregnancy‑associated disorder characterized by increased oxidative stress and inflammation. STAT3 signaling plays a key role in modulating important processes such as cell proliferation, differentiation, invasion and apoptosis. The present review aimed to analyse the role of STAT3 signaling in PE pregnancies, discuss the main natural and synthetic compounds involved in modulation of this signaling both in vivo and in vitro and summarize the main cellular modulators of this signaling to identify possible therapeutic targets and treatments to improve the outcome of PE pregnancies.

Introduction

The placenta is a transitory but key organ that undergoes changes during pregnancy ensuring the normal fetal development (1,2). Placental development is a process tightly regulated during pregnancy and its alteration can lead to complications such as preeclampsia (PE) (3), fetal growth restriction (FGR) (4), gestational trophoblastic disease (5), preterm delivery (6) and gestational diabetes mellitus (GDM) (7). In addition, placental development is impaired by exposure to exogenous agents such as bacteria (8), viruses (9) and pollutants (10,11) that can alter the normal placental function.

Preeclampsia (PE) is a hypertensive disorder of pregnancy with incidence between 2 and 10% of pregnancies worldwide (12). It is generally diagnosed from the second trimester of gestation and is clinically characterized by de novo maternal hypertension (diastolic blood pressure of 90 mmHg and/or systolic blood pressure of 140 mmHg) and proteinuria (>300 mg/24 h) (13,14). A high body mass index, previous preeclamptic pregnancy, advanced maternal age and nulliparity are important risk factors of PE (15,16).

Although clinical diagnosis of PE occurs after 20 weeks of pregnancy, it is hypothesized that placental impairment begins during early stage of pregnancy and may be due to poor trophoblast invasion (12). Poor endometrial invasion of the extravillous trophoblast (EVT) into the maternal uterine wall impairs proper remodelling of endometrium spiral arteries, altering vascular perfusion and leading to a hypoxic environment. This causes increased oxidative stress and inflammation that leads to trophoblast immaturity and altered angiogenesis of placental villi (17,18).

Depending on the gestational age of occurrence of clinical signs and symptoms, PE is divided into late-(≥34 weeks of gestation) and early-onset PE (<34 weeks of gestation). Late-onset PE accounts for the majority of preeclampsia cases (~90%), while early-onset PE is less common but associated with higher rates of neonatal mortality and a greater degree of maternal morbidity compared with late-onset PE (19,20).

Late-onset PE is a serious condition since it can lead to eclampsia and hemolysis, elevated liver enzyme and low platelet syndrome (20,21). Early-onset PE is associated with impaired remodelling of the uterine spiral arteries, which leads to hypoxia, trophoblast immaturity, maternal systemic inflammation, vascular dysfunction and FGR (18,22,23) while late-onset PE is associated with maternal endothelial dysfunction (19,22). Both late- and early-onset PE show increased oxidative stress and inflammatory response that can cause maternal and fetal complications (19,20,24). Thus, early- and late-onset PE are considered distinct forms of PE with different pathophysiology and pregnancy outcomes.

Numerous studies (25-27) have observed involvement of the immune system in the development of PE. Moreover, the cytokine environment serves a pivotal role in the differentiation of T cell subsets (Fig. 1). T helper (Th) 1 cells are induced by IFN-γ and IL-12, two inflammatory mediators involved in the activation of the adaptive immune response (28). Th2 cell differentiation is induced by IL-4 while Th17 cell differentiation is induced in presence of IL-6 and transforming growth factor-β (TGF-β). TGF-β is also responsible for T regulatory (Treg) cell differentiation, an important T cell subset in maintaining self-tolerance and pregnancy (29,30). Th17 cells are considered the key effector T cells in induction of the inflammatory response (28,30).

PE pregnancies show an imbalance between inflammatory Th1/Th17/Th2 and Treg profiles (30,31). In PE pregnancies, CD4+ T cell subsets have a high expression of transcription factor T box (T-bet) and retinoic acid receptor-related orphan receptor γ (RORγt), which are characteristic of Th1 and Th17 profiles, respectively, and decreased expression of GATA 3 binding protein (GATA-3), and forkhead box P3 (FoxP3), associated with Th2 and Treg profiles, respectively (32). Thus, PE pregnancies have a higher percentage of Th17 cells, while levels of Treg cells are lower, suggesting a shift from Treg toward Th17 cells in PE (33). PE pregnancies are also characterized by a shift from Th2 cells toward Th1 cells (34). An imbalance of T cell subsets can alter the immune microenvironment, leading to pregnancy complications, including PE (30,33).

Signal transducer and activator of transcription 3 (STAT3) is a signal transducer protein activated by the binding of cytokine or growth factors to their specific receptors. STAT3 serves a key role in regulating immune response by modulating Th1/Th17/Th2 and Treg gene expression profiles (35).

STAT3 is key for embryonic development: STAT3-deficient embryos die in utero; transient suppression of STAT3 notably decreases implantation and suppresses decidualization, demonstrating a key role of STAT3 in embryo implantation and development (36). Moreover, STAT3 regulates important trophoblast cell processes such as proliferation, migration, differentiation and apoptosis (37,38). Since these processes are altered in PE pregnancies (18,39-41), STAT3 signaling may play a key role in this pathology.

The aim of the present review is to provide an overview of the role of STAT3 signaling in PE pregnancy and understand the potential therapeutic use of STAT3 modulators to provide new therapeutic approaches in treatment and management of PE.

STAT3 signaling

STAT family of proteins comprises seven transcription factors: STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b and STAT6. STAT3 has 770 amino acids and is organized in six domains (Fig. 2). The N-terminal domain is involved in interaction with other co-activators [such as c-Jun and CREB binding protein (CBP) p300]; the coiled-coil domain is necessary for the binding to the activated receptor, the DNA-binding domain is involved in recognizing the target DNA consensus sequence, the SH2 domain is necessary for STAT3 dimerization and recognizes the phosphorylated (p) tyrosine motifs (Y705 residue) in the STAT3 trans-activation domain (which contains conserved Tyr and Ser residues). STAT family members can form heterodimers (with another member of the STAT protein family) or homodimers (with another identical STAT protein) to initiate transcriptional activity of STAT-dependent genes following ligand stimulation (42,43).

When cytokines or growth factors bind to cell membrane receptors, STAT3 protein is activated (phosphorylated) by Janus kinase (JAK) family proteins, which are receptor-associated tyrosine kinases. Following phosphorylation, STAT3 forms a homodimer that is transferred into the nucleus via the nuclear pore complex (a GTP-dependent process) to bind the base sequence TTCnnGAA in the promoter of STAT3-dependent genes, thereby activating their transcription (44). STAT3 is also phosphorylated by non-receptor tyrosine kinases such as SRC and ABL (45,46).

STAT3 long-term activation is inhibited by suppressors of cytokine signaling (SOCS) protein, creating a negative feedback loop (44). STAT3 signaling is also inhibited by the protein inhibitor of activated STAT, which blocks the DNA-binding activity of STAT3, inhibiting its transcriptional activity (44). STAT3 signaling serves a key role in several processes including cell proliferation, migration, survival and differentiation (Fig. 3) (42,44,47).

STAT3 signaling in PE

STAT3 serves a key role in the transcriptional activation of several proteins involved in the regulation of numerous cell processes such as apoptosis, cell proliferation and invasion (48,49). In trophoblast cells, STAT3 inhibition induces apoptosis and decreases cell proliferation and invasion, key processes involved in normal placental development (50-52). Thus, decreased STAT3 expression may inhibit EVT invasion into the maternal uterine wall, causing shallow trophoblast invasion and placental malperfusion, which are characteristics of PE pregnancy. These effects of STAT3 on trophoblast cells may be due to modulation of MMP activity since it has been demonstrated that STAT3 activation inhibits tissue inhibitor of metalloproteinase (TIMP)-1 expression (51).

STAT3 and pSTAT3 expression is significantly lower in human PE placentas compared with normal placentas (53-60). Thus, decreased STAT3 expression and activation in PE placental tissues serve an important role in the pathogenesis of PE. However, in vivo models of PE show contrasting results: To the best of our knowledge, there is only one study that reflects results obtained in humans (61). Other studies showed an increased pSTAT3 expression in placentas of PE models (62-65) while another study showed no alteration in pSTAT3 expression (66). Thus, studies focused on the role and modulation of STAT3 in in vivo models of PE must always be assessed for pSTAT3 expression to reflect the data found in humans.

Soluble Flt-1 (sFlt-1), also called soluble vascular endothelial growth factor (VEGF) receptor 1, is a soluble form of the VEGF and placental growth factor (PLGF) receptor, and plays a key role in decreasing levels of free PLGF and VEGF, causing endothelial dysfunction (67). sFlt-1 levels are increased in PE and associated with PE severity (68,69). Another important anti-angiogenic factor is the soluble endoglin (sEng), a co-receptor for TGFβ-1 and TGFβ-3, produced by proteolytic cleavage of endoglin and involved in angiogenesis and endothelial cell differentiation. sEng serves a key role in PE pathogenesis since it decreases circulating levels of TGFβ, a key modulator of angiogenesis (70), inhibiting TGFβ pathway and leading to endothelial dysfunction (71). Although the placenta is the primary source of these circulating factors, it has been demonstrated that peripheral blood mononuclear cells (PBMCs) may be an additional source of sFlt-1 and sEng (72).

A previous study (73) showed that STAT3 mRNA and protein levels are increased in CD4+ and CD8+ T lymphocytes isolated from patients with PE compared with those from normal pregnancies. The aforementioned study found an increase in Th17 lymphocytes while Treg population was notably lower in PE, demonstrating an increased Th17/Treg ratio in PE. Furthermore, levels of cytokines (IL-17 and IL-22) and anti-angiogenic molecules such as sEng and sFlt-1 are increased in isolated CD4+ cells from patients with PE, suggesting a possible association with PE pathogenicity. STAT3 is required for the IL-17 production by Th17, a subtype of CD4+ T lymphocytes (74). Thus, CD4+ T cells could participate in production of antiangiogenic factors characterising PE pregnancy. In particular, STAT3 could participate in favoring hypertension development during PE by increasing IL-17 levels, which serves an important role in hypertension (75).

STAT3 signaling serves a key role in angiogenesis since the inhibition of STAT3 phosphorylation suppresses this process (76). Normally, pSTAT3 is strongly expressed in the endothelial cells of first and second trimester placentas but its expression notably decreases in the third trimester, when angiogenesis is completed (77). However, pSTAT3 expression is notably increased in endothelial cells of PE placentas compared with normotensive controls, suggesting an association between STAT3 activation and placental angiogenesis defects in PE (78).

Placental factors in maternal circulation cause systemic endothelial dysfunction in PE pregnancy, increasing the risk of cardiovascular disease (CVD) following delivery (79-81). Christensen et al (82) found that human umbilical vein endothelial cell treatment with serum from patients with PE notably decreases STAT3(Y705) phosphorylation compared with serum from uncomplicated pregnancy. Moreover, sera from patients with previous PE, current hypertension and carotid atherosclerotic plaques shows significantly lower STAT3(Y705) phosphorylation capabilities compared with healthy controls with previous uncomplicated pregnancies 8-18 years after delivery. Thus, decreased serum-induced endothelial STAT3(Y705) activation may play an important role in PE-associated endothelial dysfunction and reduced endothelial STAT3(Y705) phosphorylation and may increase post-preeclamptic CVD risk after delivery (82).

Decreased STAT3 expression in PE placentas may favor trophoblast apoptosis in the hypoxic environment at the beginning of placentation (50-52). Moreover, decreased expression of STAT3 is associated with reduced cell proliferation and invasion due to decreased activity of MMPs (51). Decreased STAT3 expression can inhibit EVT invasion into the maternal uterine wall, causing a shallow trophoblast invasion, which is one of the primary causes of PE occurrence (12). Since STAT3 expression is significantly increased in CD4+ and CD8+ T lymphocytes isolated from patients with PE (73), STAT3 pathway is also involved, at least in part, in the increased inflammatory cytokines found in serum of patients with PE (83). Therefore, investigating the effects of STAT3 signaling modulation in PE pregnancy is necessary to understand and treat this complication of pregnancy.

STAT3 modulation in PE by natural and synthetic compounds

STAT3 modulation by natural compounds

Natural compounds, also known as phytotherapeutics, are biological substances found in several plants, bacteria, fungi and marine organisms and are used worldwide due to anti-inflammatory, antioxidant and anticancer effects (84-87).

Silibinin is a flavonolignan derived from milk thistle (Silybum marianum) with antioxidant and anticancer properties (88). Xu et al (89) demonstrated that silibinin treatment of PBMCs, isolated from patients with PE pregnancy, decreases STAT3/RORγt expression, which is involved in the regulation of Th17 inflammatory profiles. In addition, PBMCs treated with silibinin show lower concentrations of inflammatory cytokines such as TNF-α, IL-6, and IL-23 and higher levels of IL-10 and TGF-β. Since there is a shift from Treg cells toward Th17 cells in PE pregnancy (33), silibinin may be an important immunomodulatory compound able to regulate the Th17/Treg cell balance in PE, decreasing inflammation (90).

Paeonol is a natural compound extracted from Cortex Moutan, a plant used in Chinese medicine with anti-inflammatory, anticancer and antioxidant effects (91,92). Paeonol significantly attenuates the inflammatory response in the placenta of PE mouse model, decreasing mRNA levels of TNF-α, IL-6 and IFN-γ and increasing IL-4 mRNA levels. Furthermore, paeonol treatment inhibits phosphorylation of JAK2 and STAT3 in the placental tissues of PE mouse model (62). These effects are reversed by treatment with SC-39100, a JAK2/STAT3 pathway agonist, demonstrating that paeonol anti-inflammatory effects are due to inhibition of the JAK2/STAT3 signaling pathway (62).

Vitamin D is a pro-hormone primarily obtained from exposure of the skin to ultraviolet B radiation and can regulate inflammatory responses, favouring the shift from a pro-inflammatory to a tolerogenic immune response by modulating numerous cells of the immune system including CD4+ T cells (93-95). Vitamin D deficiency may contribute to PE onset, altering Th1/Th17 and Th2/Treg profiles (96). Ribeiro et al (97) found that plasma levels of vitamin D are lower in PE compared with normal pregnancies. Vitamin D treatment of CD4+ T cells isolated from PE decreases STAT1/STAT4/T-bet and STAT3/RORγt activation, while increasing STAT6/GATA-3 and STAT5/FoxP3 activation. Treatment of PBMCs isolated from patients with PE with vitamin D also decreases levels of IFNγ, TNFα, IL-17, IL-22, IL-23 and IL-6, while increasing IL-10 and TGF-β levels, suggesting an immunomodulatory effect of vitamin D on STAT signaling that favours the shift from Th1/Th17 to Th2/Treg profiles. Vitamin D treatment may exert a beneficial effect in ameliorating systemic inflammation characterising PE pregnancies (97).

STAT3 modulation by synthetic compounds

Studies (98-100) have reported that STAT3 can also be regulated by synthetic compounds. SO2, a major air pollutant produced by industrial factories and vehicle exhaust, can lead to pregnancy complications such as stillbirth, preterm birth, GDM and PE (101-104). Treatment of Swan.71 trophoblast cells with SO2 derivatives significantly decreases cell migration and invasion, arrests the cell cycle at S/G2/M phase and induces apoptosis (105). Moreover, SO2 derivatives notably increase IL-1β and decrease IL-6 secretion and STAT3 phosphorylation, leading to decreased expression of MMP2 and MMP9; this indicates SO2 derivatives exert their toxic effects on trophoblast cells, inhibiting IL-6/STAT3 signaling, which plays a key role in regulating cell viability, invasion and migration (105).

Pravastatin is a statin normally used to treat CVD but it can also reduce cholesterol content, alleviate inflammation, decrease oxidative stress and regulate endothelial functions (106,107). Wang et al (63) found that the expression of serum IL-6 in PE rat model (obtained by deoxycorticosterone acetate injection) is markedly higher than in controls and significantly reduced in PE rats treated with pravastatin. Moreover, PE rats show increased expression of pSTAT3 compared with controls rats. However, pSTAT3 expression is significantly decreased in PE rats treated with pravastatin. The proliferation of rat trophoblast cells is significantly decreased in PE rats (due to increased apoptosis) compared with controls but significantly increased in PE rats treated with pravastatin (which reduced apoptosis), indicating that pravastatin can inhibit IL-6/STAT3 signaling, decreasing the apoptosis of trophoblast cells in PE rats (63). These data are in contrast with another study that evaluated the role of pravastatin in a PE-like mice model (obtained by adenoviral overexpression of sFlt-1), which showed high blood pressure, abnormal vascular reactivity and proteinuria similar to PE (108,109). However, pSTAT3 placental expression is significantly higher in PE-like mice treated with pravastatin compared with untreated and control (normal) groups, suggesting that pravastatin can prevent PE and modulate STAT3 activation, further validating a beneficial role of statins in preventing PE (66). The contrasting results may be due to the different PE models.

Sulfasalazine is an anti-inflammatory drug used to treat arthritis and inflammatory bowel disease (110,111). However, sulfasalazine decreases placental secretion of the anti-angiogenic factor sFlt-1, expression of which is regulated by the epidermal growth factor receptor (EGFR) signaling pathways (112). By using primary cytotrophoblast cells, Hastie et al (113) found that sulfasalazine decreases sFlt-1 secretion, downregulating EGFR expression. Additionally, sulfasalazine notably decreases protein expression of ERK1/2 and STAT3, which are key adaptor molecules downstream of EGFR. Thus, sulfasalazine decreases sFlt-1 secretion, downregulating EGFR/ERK and EGFR/STAT3 signaling (113).

Nω-nitro-L-arginine methyl ester (L-NAME) is a L-arginine analogue used as a nitric oxide synthase inhibitor to treat hypotension. Due to these effects, it is also used to establish the PE-like rat model (114). L-NAME administration causes a decreased expression of STAT3 and pSTAT3 in the placenta of PE rats, suggesting a role of STAT3 signaling in the development of PE (61). Rats treated with L-NAME are widely used as an in vivo model to study PE pathophysiology (115-118).

Montelukast is a drug used in chronic asthmatic patients planning for pregnancy and is safely used during pregnancy (119). Montelukast is a selective cysteinyl leukotriene receptor antagonist with antioxidant and anti-inflammatory effects (120,121). Abdelzaher et al (64) demonstrated that montelukast treatment decreases oxidative stress and expression of IL-6, TNF-α, pJAK2, and STAT3 in PE rats. Thus, montelukast exerts anti-inflammatory effects, suppressing the IL-6/JAK2/STAT3 signaling pathway in PE rats (Table I) (64).

Table I

STAT3 modulation by natural and synthetic compounds.

Table I

STAT3 modulation by natural and synthetic compounds.

A, Natural compounds
ModulatorModelResults(Refs.)
SilibininPBMCDecreases STAT3/RORγt, TNF-α, IL-6 and IL-23 levels; increases IL-10 and TGF-β expression(90)
PaeonolPE mouse model (induced by PS/PC injection)Decreases TNF-α, IL-6, and IFN-γ mRNA levels; increases IL-4 mRNA levels; inhibits phosphorylation of JAK2 and STAT3; effects are reversed by treatment with SC-39100, a JAK2/STAT3 pathway agonist.(62)
Vitamin DPBMC and CD4+ T cells from patients with PEDecreases STAT1/STAT4/T-bet and STAT3/RORγt signaling and expression of IFNγ, TNFα, IL-17, IL-22, IL-23 and IL-6; increases STAT6/GATA-3 and STAT5/FoxP3 signaling and expression of IL-10 and TGF-β(97)

B, Synthetic compounds

ModulatorModelResults(Refs.)

SO2 derivatesSwan.71 trophoblast cellsDecreases cell migration and invasion; arrests cell cycle at S/G2/M phase; induces apoptosis; increases IL-1β secretion; decreases IL-6 secretion, STAT3 phosphorylation and MMP2 and MMP9 expression(105)
PravastatinPE rat model (induced by deoxycorticosterone acetate injection)Decreases IL-6, pSTAT1 and pSTAT3 expression(63)
PE mouse model (induced by sFlt1 overexpression)Increases pSTAT3 expression in placenta(66)
SulfasalazinePrimary cytotrophoblast cellsDecreases sFlt-1 secretion and protein expression of ERK1/2 and STAT3(113)
L-NAMEPE rat model (induced by L-NAME)Decreases sSTAT3 and pSTAT3 expression in the placenta(61)
MontelukastPE rat model (induced by L-NAME)Decreases oxidative stress and expression of IL-6, TNF-α, pJAK2 and STAT3 in placental tissues(64)

[i] PS/PC, phosphatidylserine/dioleoyl-phosphatidylcholine; L-NAME, Nω-nitro-L-arginine methyl ester; PE, preeclampsia; p, phosphorylated; T-bet, T-box protein.

STAT3 modulation in PE by non-coding (nc)RNAs

ncRNAs are functional RNA molecules without protein-coding abilities. The most studied ncRNAs include microRNA (miRNA or miR), long ncRNA (lncRNA) and circular RNA (circRNA) (122-124). ncRNAs regulate expression of genes involved in several cellular processes including cell proliferation, invasion and metabolism (122).

miRNAs are a group of endogenous small single-strand ncRNAs that exert multifaceted functions in numerous diseases (123-125). The miR-133 family (comprising miR-133a and miR-133b) can affect invasion, migration and proliferation of tumor cells (126) and plays a key role in pregnancy complications such as recurrent spontaneous abortion (127). Placental tissue of patients with PE shows high levels of miR-133b and decreased pJAK2 and pSTAT3 expression. In HTR8/SVneo cells, hypoxia induces miR-133b expression and decreases pJAK2 and pSTAT3 expression and trophoblast migration and invasion while increasing apoptosis, thereby proving that miR-133b may exert its functions by regulating the JAK2/STAT3 pathway (56). Thus, inhibiting miR-133b may improve oxidative stress injury (induced by hypoxia) to promote the migration and invasion of trophoblasts and suppress apoptosis by activating the JAK2/STAT3 pathway (56).

Another miRNA involved in STAT3 modulation and altered in PE is miR-125b. This miRNA is associated with extra-villous trophoblastic proliferation and invasion, and its expression is notably increased in PE (128). Serum levels of miR-125b are significantly increased in patients with PE compared with normal pregnancies (128). Moreover, high levels of miR-125b decrease HTR-8/SVneo cell proliferation, invasion and migration as well as expression of STAT3, pSTAT3 and SOCS3, demonstrating that STAT3 is a target gene of miR-125b; high levels of miR-125b inhibit STAT3 signaling, reducing migration and invasion of extra-villous trophoblast cells (129).

In addition to miRNAs, several studies have demonstrated a key role of lncRNAs in pregnancy complications such as PE (130) and GDM (131). Dendritic cells (DCs) serve a key role as primary antigen-presenting cells at the beginning of pregnancy and express lnc-DC, which induces DC differentiation, maturation and STAT3 phosphorylation (132). Pregnancy complications characterized by impaired trophoblast invasion such as PE exhibit excessive DC maturity (133). Moreover, it has been reported that expression of lnc-DC and pSTAT3 is increased in the decidua of patients with PE (134). Furthermore, the proportion of Th1 cells and mature DCs is notably higher in patients with PE, suggesting that upregulation of lnc-DC induces the over-maturation of decidual DCs in PE, leading to an increase in Th1 cells that lead to a persistent inflammatory response (134). Overexpression of lnc-DC in HTR8/SVneo cells inhibits trophoblast invasion and motility by increasing pSTAT3 levels and TIMP1 and 2 expression while decreasing expression of MMP-9, -2 and -3 (135). Thus, lnc-DC regulates trophoblast invasion and motility by modulating STAT3 activation and MMP expression (135).

circRNAs are single-strand ring-like ncRNAs produced by reverse splicing of precursor RNA following transcription and are involved in numerous pathologies including pregnancy complications (136). Expression of circRNA of pregnancy-associated plasma protein A (circPAPPA) is downregulated in PE. Knockdown of circPAPPA in HTR8-S/Vneo cells decreases proliferation and invasion ability. Moreover, miR-384 (which targets STAT3) is a direct target of circPAPPA (57). STAT3 expression is decreased when circPAPPA is knocked down, suggesting that downregulation of circPAPPA facilitates onset and development of PE by suppressing trophoblast invasion and proliferation via modulation of miR-384/STAT3 signaling (Table II) (57).

Table II

STAT3 modulation in HTR8/SVneo cells by ncRNAs.

Table II

STAT3 modulation in HTR8/SVneo cells by ncRNAs.

ModulatorResults(Refs.)
miR-133bDecreases pJAK2 and pSTAT3 expression and trophoblast migration and invasion; induces apoptosis(56)
miR-125bDecreases cell proliferation, invasion and migration and expression of STAT3, pSTAT3 and SOCS3(129)
lnc-DCInhibits cell invasion and motility; increases pSTAT3 and TIMP-1 and -2 expression; decreases MMP-9, -2 and -3 expression(135)
circPAPPAKnockdown of circPAPPA decreases cell proliferation and invasion and STAT3 expression by increasing miR-384 expression(57)

[i] TIMP, tissue inhibitor of metalloproteinase; lnc, long non-coding; miR, microRNA; p, phosphorylated; SOCS, suppressor of cytokine signaling proteins; DC, dendritic cell; circPAPPA, circular pregnancy-associated plasma protein A RNA.

Cellular STAT3 modulation in PE

B7-H4 is a type I transmembrane glycoprotein belonging to the B7 family of immune checkpoint proteins, which are involved in regulation of immune response, preventing its excessive activation (137). B7-H4 is normally expressed in the placental villous but its expression significantly decreases in decidua of patients with PE compared with normal controls (138). Contrarily, B7-H4 serum levels are notably increased in patients with PE (139). B7-H4 treatment of SGHPL-5 trophoblast cells inhibits proliferation, migration and invasion while promoting apoptosis by decreasing pAKT, pPI3K and pSTAT3 expression (140). Thus, B7-H4 may serve an important role in shallow trophoblast invasion in PE.

RAR-related orphan receptor A (RORA) is a member of the ROR subfamily that serves as a transcription factor in the regulatory region of ROR-responsive genes; its expression is induced by hypoxia (141,142). It has been reported that RORA expression is increased in PE tissues but also in HTR-8/SVneo cells exposed to hypoxic conditions. Silencing of RORA in HTR-8/SVneo cells increases migration and proliferation while decreasing pSTAT3 and pJAK2 expression. The inhibitory effects of RORA silencing are reversed when cells are treated with the STAT3 activator RO8191 (143). Thus, RORA regulates trophoblast cell proliferation and migration via the JAK2/STAT3 signaling pathway (143).

Basal cell adhesion molecule (BCAM) belongs to the immunoglobulin superfamily and is involved in cellular processes such as cell adhesion, migration and invasion (144). Liu et al (58) found that BCAM expression is significantly decreased in PE placenta. Moreover, silencing BCAM in HTR-8/SVneo and JAR cells leads to decreased trophoblast proliferation, migration and invasion and suppresses pSTAT3(Y705) expression through the downregulation of phosphoinositide-3-kinase regulatory subunit 6 (PIK3R6) expression, a kinase that phosphorylates STAT3. However, phosphorylation on S727 of STAT3 is not altered by BCAM deficiency. In addition, adenoviruses containing BCAM short hairpin RNA genes (Ad-shBCAM) cause BCAM deficiency and a PE-like phenotype with elevated systolic blood pressure, proteinuria and FGR. Accordingly, the expression of BCAM, PIK3R6 and pSTAT3 is downregulated in Ad-shBCAM rats (58). Thus, BCAM deficiency decreases trophoblast proliferation, migration and invasion by inhibiting PIK3R6/STAT3 signaling (58).

NADPH oxidase 2 (Nox2) is an important source of ROS and serves a key role in ferroptosis (145,146), a type of programmed cell death due to iron-dependent lipid peroxidation (147). STAT3 serves as an oxidation-responsive transcription factor and plays a key role in regulating ferroptosis as it can bind the promoter region of ferroptosis-associated genes such as glutathione peroxidase 4 (GPX4) (148,149). A previous study (59) found that Nox2 expression is significantly higher in PE placentas while STAT3 and GPX4 expression is decreased. Moreover, STAT3 and GPX4 gene expression is notably decreased by hypoxia and RAS-selective lethal compound 3 (RLS3)-induced ferroptosis while their expression is restored when ferroptosis is inhibited with Ferrostatin-1 (Fer-1). Silencing Nox2 in HTR8/SVneo cells inhibits ferroptosis and increases STAT3 and GPX4 expression (59). Thus, Nox2 may trigger ferroptosis in PE via modulation of the STAT3/GPX4 pathway (59).

NOP2/Sun5 (NSUN5) is an RNA methyltransferase involved in important cell processes such as mitochondria assembly and cell proliferation (150). Zhang et al (151) found a notable association between NSUN5 polymorphism (rs77133388) and PE. Pregnant single-base mutant mice (NSUN5 R295C at rs77133388) exhibit PE symptoms and reduced decidualization. Additionally, the aforementioned study found decreased IL-11Rα, cyclin D3, pJAK2 and pSTAT3 expression in NSUN5 R295C mice, suggesting that NSUN5 mutation potentially alters decidualization through IL-11Rα/JAK2/STAT3/cyclin D3 signaling, favouring PE occurrence (151).

Hypoxia-inducible factors (HIFs; HIF-α and HIF-β subunits) are hypoxia-induced transcription factors that regulate cellular processes under hypoxic condition. HIF-1α, HIF-2α and HIF-3α are paralogs of the HIF-α subunit; while HIF-1α and HIF-2α are considered the two master regulators of hypoxic response, little is known about HIF-3α (152). Qu et al (153) found that HIF-3α expression is decreased in PE placentas compared with normal pregnancy. Moreover, under chronic hypoxia (72 h), the expression of HIF-3α, pJAK and pSTAT3 is significantly decreased while apoptosis notably increases. Overexpression of HIF-3α in HTR8/SVneo cells markedly increases phosphorylation of JAK/STAT, indicating that HIF-3α upregulation can regulate the JAK/STAT pathway and serves as a protective factor against hypoxia, favoring cell survival (153).

Annexin A1 (ANXA1) is a calcium-dependent phospholipid-binding protein that can bind negatively charged phospholipids and is involved in cell activities such as anti-inflammatory response, differentiation and proliferation, cell signal regulation and phagocytosis of apoptotic cells (154-156). ANXA1 is highly expressed in the plasma of patients with PE (157,158). Feng et al (159) found that ANXA1, TNF-α, IL-1β, IL-6 and IL-8 expression are increased in placental tissues of PE rats. In addition, the aforementioned study found that silencing of ANXA1 in trophoblast cells isolated from placentas of PE rat model significantly decreases apoptosis and inflammatory response of trophoblast cells. Furthermore, silencing of ANXA1 significantly increases expression of Bcl-2 and pro-caspase-3, while downregulating the expression of BAX, cleaved-caspase-3, TNF-α, IL-1β, IL-6 and IL-8 (159). Silencing of ANXA1 notably decreases phosphorylation of JAK2 and STAT3 (159). These effects on STAT3 and JAK2 can be explained by an indirect modulation by ANXA1. Decreased phosphorylation of JAK2 and STAT3 may be due to decreased expression of the cytokines TNF-α, IL-1β, IL-6 and IL-8, which activate JAK/STAT3 pathway (160-163). Similar results were obtained by Mo et al (164) studying ANXA7, another member of the annexin family (154). Silencing of ANXA7 in HTR-8/SVneo cells induces cell apoptosis and inhibits cell proliferation by downregulating Bcl-2 protein expression (164). Silencing of ANXA7 decreases pJAK and pSTAT3 expression (164). As it has been reported that trophoblast viability is notably decreased when the levels of pJAK and pSTAT3 are reduced (165), the aforementioned results demonstrated that ANXA7 can regulate trophoblast apoptosis by modulating the JAK/STAT3 pathway.

IL-27 is a member of the IL-12/IL-6 family of cytokines produced by antigen-presenting cells and regulates T cell differentiation and function, exerting pro- and anti-inflammatory effects during immune response (166). Expression of IL-27 and its receptor (IL-27 receptor α) is significantly increased in the trophoblast of placentas from PE pregnancy (167). A previous study found that IL-27 significantly inhibits HTR-8/SVneo cell invasion and migration by favouring the expression of epithelial markers over mesenchymal markers. Furthermore, IL-27 induces phosphorylation of STAT1 and STAT3 (168). Silencing of STAT1 attenuates the effects of IL-27, while silencing STAT3 has no effect, demonstrating that IL-27 may inhibit trophoblast cell migration and invasion by affecting epithelial-mesenchymal transition via a STAT1-dominant pathway in PE (168).

Heme oxygenase-1 (HO-1) is a key antioxidant enzyme with anti-hypertensive effects (169) and cytoprotective and anti-inflammatory functions under ischemic conditions (170). HO-1 expression is significantly increased in PE placentas while STAT3 phosphorylation (Y705) is notably decreased. Moreover, human placental choriocarcinoma JEG-3 cells exposed to hypoxia show increased HO-1 expression and STAT3 phosphorylation (Y705) compared with cells cultured under normoxic conditions (60). HO-1 overexpression in JEG-3 cells significantly inhibits hypoxia-promoted STAT3 phosphorylation (Y705), suggesting that the overexpression of HO-1 in PE placentas might contribute to decreased STAT3 phosphorylation (Y705) found in the placentas complicated by PE (60). This is consistent with a study by Xu et al (165), reporting an increased expression of pJAK and pSTAT3 in primary third trimester trophoblast cells exposed to hypoxia. To the best of our knowledge, the aforementioned study is the only report of increased expression of total STAT3 expression in PE placentas.

HO-1 expression and activity are induced by cobalt protoporphyrin (CoPP) (171) and HO-1 induction notably attenuates oxidative stress and hypertension in pregnant rats with reduced uterine perfusion pressure (RUPP) (172). A previous study found that phosphorylation of JNK, STAT1, pSTAT3 (Y705) is significantly increased in placental tissues of RUPP rats (65). CoPP decreases RUPP-induced phosphorylation of JNK and STAT1, while increasing phosphorylation of STAT3, indicating that RUPP induces oxidative stress, increasing phosphorylation of mediators of cell death, such as STAT1 and JNK (172,173), and survival, such as STAT3 (174), in placentas of pregnant rats. HO-1 induction by CoPP shifts this balance to a pro-survival phenotype by increasing phosphorylation of the pro-survival STAT3, while suppressing phosphorylation of JNK and STAT1. These results demonstrate therapeutic activity of HO-1 induction in placental cell following ischemic injury (due to the RUPP model) favouring the survival of placental cells. Thus, the HO-1 pathway may be a promising therapeutic target for management of PE (65).

Ribosomal protein S4, Y-linked 1 (RPS4Y1) is a member of the S4E family of ribosomal proteins ubiquitously expressed and is involved in regulation of cell processes such as apoptosis, cell migration and invasion (175,176). A previous study (177) found that RPS4Y1 levels are significantly upregulated in PE placentas. Silencing of RPS4Y1 in HTR8/SVneo cells induces cell invasion and increased STAT3 phosphorylation along with increased expression of N-cadherin and vimentin. These effects are abolished when RPS4Y1 and STAT3 are silenced, demonstrating that RPS4Y1 may be involved in PE, affecting trophoblast cell migration and invasion via the STAT3 pathway (177).

EGF is a polypeptide involved in cell proliferation, differentiation and survival (178). Lower levels of EGF are found in plasma and urine of patients with PE, suggesting a potential role of EGF in this pathology (179). A previous study (180) reported that treatment of HTR-8/SVneo cells with EGF notably increases cell invasion. Moreover, EGF treatment leads to an increase in phosphorylation of ERK1/2, STAT1 and STAT3 (at both Y705 and S727 residues). Inhibition of ERK1/2 phosphorylation by U0126 decreases EGF-mediated invasion and pSTAT3 and pSTAT1 expression. Silencing of STAT3 leads to decreased EGF-mediated invasion of HTR-8/SVneo cells and pSTAT1 expression but does not have any effect on ERK1/2 activation. Silencing of STAT1 also leads to decreased EGF-mediated invasion of HTR-8/SVneo cells and ERK1/2 and STAT3 (at S727 residue) phosphorylation (180). These results suggest crosstalk between ERK1/2 and JAK/STAT pathways during EGF-mediated increase of HTR-8/SVneo cells invasion; phosphorylation at S727 residue of both STAT3 and STAT1 may be critical in this process (180).

Human leucocyte antigen-G (HLA-G) is the primary immune modulator in embryo implantation and allows the interaction between immune cells [such as natural killer (NK) cells] and trophoblast cells, inhibiting NK cytotoxicity and cytokine production (181). Moreover, decreased HLA-G expression may contribute to PE onset (182). Silencing HLA-G in JEG-3 cells decreases invasion capacity but does not alter cell proliferation or apoptosis. Moreover, silencing of HLA-G decreases STAT3 activation, whereas the overexpression of HLA-G promotes STAT3 activation and invasion in JEG-3 cells, demonstrating that HLA-G is able to regulate JEG-3 cell invasion by influencing STAT3 activation explaining implantation defects due to the low HLA-G expression in PE (Table III) (183).

Table III

Cellular STAT3 modulation in PE.

Table III

Cellular STAT3 modulation in PE.

ModulatorModelResults(Refs.)
B7-H4SGHPL-5 cellsInhibits cell proliferation, migration, and invasion; promotesapoptosis; downregulates pPI3K, pAkt and pSTAT3 expression(140)
RORAHTR-8 cellsRORA silencing increases cell migration and proliferation and decreases pSTAT3 and pJAK2 expression; STAT3 activator RO8191 reverses the inhibitory effects of RORA silencing(143)
BCAMHTR-8/SVneo, JAR cellsBCAM silencing decreases cell proliferation, migration and invasion, as well as pSTAT3(Y705) expression via the downregulation of PIK3R6 but does not alter pSTAT3(S727) expression(58)
Nox2HTR-8/SVneo cells and placental tissueNox2 expression is increased in PE; Nox2 silencing inhibits ferroptosis and increases mRNA and protein levels of STAT3 and GPX4(59)
NSUN5Patients with NSUN5 R295C (rs77133388)NSUN5 R295C decreases decidualization and IL-11Rα, cyclin D3, pJAK2 and pSTAT3 expression(151)
HIF-3αHTR8/SVneo cellsHIF-3α overexpression increases Flt1 expression and phosphorylation of JAK/STAT pathway proteins(153)
ANXA1Trophoblast cells isolated from PE rats (induced by L-NAME)ANXA1, TNF-α, IL-1β, IL-6 and IL-8 expression increases in placental tissue of PE rats; ANXA1 silencing in trophoblast cells increases the expression of Bcl-2 and pro-caspase-3; ANXA1 silencing downregulates the expression of Bcl-2-associated X protein, cleaved-caspase-3, TNF-α, IL-1β, IL-6 and IL-8, as well as phosphorylation of JAK2 and STAT3 without altering their expression(159)
ANXA7HTR8/SVneo cellsANXA7 silencing induces apoptosis, inhibits cell proliferation and decreases phosphorylation of JAK and STAT3(Y705) without altering their expression(164)
IL-27HTR8/SVneo cellsIL-27 inhibits HTR-8/SVneo cells invasion and migration and induces phosphorylation of STAT1 and STAT3; STAT1 silencing attenuates the effect of IL-27, while silencing STAT3 has no effect(168)
HO-1PE placenta and JEG-3 cellsHypoxia increases HO-1 and pSTAT3(Y705) expression; HO-1 overexpression in JEG-3 cells inhibits hypoxia-promoted pSTAT3(Y705) expression(60)
HO-1PE mouse model (induced by RUPP)HO-1 inducer cobalt protoporphyrin increases pSTAT3 (Y705) expression(65)
RPS4Y1HTR8/SVneo cellsRPS4Y1 silencing induces cell invasion and increasespSTAT3(Y705), N-cadherin and vimentin expression; these effects are abolished when RPS4Y1 and STAT3 are silenced(177)
EGFHTR8/SVneo cellsEGF increases phosphorylation of ERK1/2, STAT1 (S727) and STAT3 (at both Y705 and S727 residues); inhibition of ERK1/2 phosphorylation by U0126 decreases EGF-mediated invasion and pSTAT3 and pSTAT1 expression; STAT3 silencing decreases EGF-mediated invasion and pSTAT1 expression but does not have any effect on ERK1/2 activation; STAT1 silencing decreases EGF-mediated invasion and ERK1/2 and STAT3 (at S727 residue) phosphorylation(180)
HLA-GJEG-3 cellsHLA-G silencing decreases STAT3 activation; HLA-G overexpression promotes STAT3 activation and cell invasion(183)

[i] RORA, RAR-related orphan receptor A; BCAM, basal cell adhesion molecule; Nox2, NADPH oxidase 2; ANXA1, annexin A1; HO-1, heme oxygenase 1; EGF, epidermal growth factor; HLA-G, human leucocyte antigen-G; RUPP, reduced uterine perfusion pressure; PE, preeclampsia; p, phosphorylated; Flt, fms-like tyrosine kinase; L-NAME, Nω-nitro-L-arginine methyl ester; RPS4Y1, ribosomal protein S4, y-linked 1; NSUN, Nol1/Nop2/SUN domain.

Others STAT3 modulators in PE

Chorionic villi serve a key role in normal placental development and function, allowing the transport of nutrients and oxygen to the foetus (184). However, the development of chorionic villi is impaired in pregnancy complications such as PE and FGR (18). Chorionic villous mesenchymal stem cells (CV-MSCs) are multipotent cells that are detached from chorionic villi and differentiate in vitro into neurocytes and hepatocytes (185). CV-MSCs serve a pivotal role in regulating trophoblast function. Chu et al (186) found that treatment of JAR, JEG-3 and HTR-8 cells under hypoxic conditions with CV-MSC supernatant markedly enhances proliferation and invasion and augments autophagy. In addition, pSTAT3 and pJAK2 levels increase following CV-MSC treatment, suggesting that CV-MSC-dependent JAK2/STAT3 signaling activation is a prerequisite for autophagy upregulation in trophoblast cells and an important factor in protecting cells from hypoxia (186).

In addition to CV-MSCs, STAT3 can also be modulated by antiphospholipid antibodies (aPLs), which have been found in patients affected by antiphospholipid syndrome (187,188). aPLs are at risk factor for recurrent miscarriage and PE onset (174) as aPL can bind the β2-glycoprotein I (β2-GPI) expressed by trophoblast cells, triggering an inflammatory response and compromising the invasiveness of trophoblast cells (190). A previous study (191) found that treatment of first trimester trophoblast cells with anti-β2-GPI monoclonal antibodies notably downregulates IL-6 secretion and pSTAT3 expression, reducing trophoblast cell invasion. Thus, aPLs limit trophoblast cell migration by downregulating trophoblast IL-6 secretion and STAT3 activation (191).

Conclusion

STAT3 signaling may be a promising target for treatment of PE. The present review summarizes the role of STAT3 signaling in regulating processes in placental cell lines and in vivo PE models. STAT3 signaling is regulated by several factors (Fig. 4). In particular, natural compounds such as silibinin, paeonol and vitamin D, as well as synthetic compounds such as SO2 derivates, pravastatin, sulfasalazine, L-NAME and montelukast, regulate STAT3 activation and/or expression in inflammatory and trophoblast cells, regulating inflammatory cytokine production and modulating trophoblast cell proliferation and invasion (61,63,64,105,113).

Furthermore, STAT3 expression is modulated by ncRNAs such as miR-133b, miR-125b, lnc-DC and circPAPPA, and cellular modulators such as RORA, BCAM, Nox2, NSUN5, IL-27, HO-1, RPS4Y1, EGF, HLA-G, ANXA1 and ANXA7. Thus, STAT3 signaling plays a key role in important processes for placental development that are impaired in PE placentas. STAT3-dependent alterations in PE may be improved by stimulating the activation of STAT3 signaling. Therapy focused on STAT3 regulation may improve the efficiency of the classical treatments (e.g. magnesium sulfate, heparin) to ameliorate PE outcomes or avoid its onset. Moreover, natural and synthetic compounds decrease pSTAT3 expression (Fig. 4). Use of these compounds must be tightly controlled or avoided since it can significantly worsen STAT3-dependent cellular and molecular processes given that pSTAT3 expression is low in PE placentas (53-55).

As STAT3 expression is notably decreased in PE placentas compared with normal placentas (53,55), its activation (especially in pregnancy at risk of PE development) may promote processes necessary for proper placental development (such as protecting trophoblast cells from apoptosis in a hypoxic environment, such as that at the beginning of placentation).

The role of STAT3 signaling in PE pregnancies has also clinical value. As STAT3 signaling is inhibited by miR-133b, miR-125b, B7-H4 and NSUN5 polymorphism (rs77133388), modulators that can be detected in the blood at first trimester of pregnancy when no PE clinical signs or symptoms are present, increased levels of these modulators in the blood or the presence of rs77133388 could be an indicator of a pregnancy at risk of PE due to a possible impairment of STAT3 signaling.

Availability of data and materials

Not applicable.

Authors' contributions

DM reviewed and analyzed the literature and wrote the manuscript. FP, NDS, SFG and AC reviewed and edited the manuscript. GT conceived the study and wrote and critically revised the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

No funding was received.

References

1 

Tossetta G, Avellini C, Licini C, Giannubilo SR, Castellucci M and Marzioni D: High temperature requirement A1 and fibronectin: Two possible players in placental tissue remodelling. Eur J Histochem. 60:27242016.

2 

Fantone S, Giannubilo SR, Marzioni D and Tossetta G: HTRA family proteins in pregnancy outcome. Tissue Cell. 72:1015492021. View Article : Google Scholar : PubMed/NCBI

3 

Marzioni D, Todros T, Cardaropoli S, Rolfo A, Lorenzi T, Ciarmela P, Romagnoli R, Paulesu L and Castellucci M: Activating protein-1 family of transcription factors in the human placenta complicated by preeclampsia with and without fetal growth restriction. Placenta. 31:919–927. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Todros T, Marzioni D, Lorenzi T, Piccoli E, Capparuccia L, Perugini V, Cardaropoli S, Romagnoli R, Gesuita R, Rolfo A, et al: Evidence for a role of TGF-beta1 in the expression and regulation of alpha-SMA in fetal growth restricted placentae. Placenta. 28:1123–1132. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Marzioni D, Quaranta A, Lorenzi T, Morroni M, Crescimanno C, De Nictolis M, Toti P, Muzzonigro G, Baldi A, De Luca A and Castellucci M: Expression pattern alterations of the serine protease HtrA1 in normal human placental tissues and in gestational trophoblastic diseases. Histol Histopathol. 24:1213–1222. 2009.PubMed/NCBI

6 

Cecati M, Sartini D, Campagna R, Biagini A, Ciavattini A, Emanuelli M and Giannubilo SR: Molecular analysis of endometrial inflammation in preterm birth. Cell Mol Biol (Noisy-le-grand). 63:51–57. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Tossetta G, Fantone S, Gesuita R, Di Renzo GC, Meyyazhagan A, Tersigni C, Scambia G, Di Simone N and Marzioni D: HtrA1 in gestational diabetes mellitus: A possible biomarker? Diagnostics (Basel). 12:27052022. View Article : Google Scholar : PubMed/NCBI

8 

Licini C, Tossetta G, Avellini C, Ciarmela P, Lorenzi T, Toti P, Gesuita R, Voltolini C, Petraglia F, Castellucci M and Marzioni D: Analysis of cell-cell junctions in human amnion and chorionic plate affected by chorioamnionitis. Histol Histopathol. 31:759–767. 2016.PubMed/NCBI

9 

Yates EF and Mulkey SB: Viral infections in pregnancy and impact on offspring neurodevelopment: Mechanisms and lessons learned. Pediatr Res. 96:64–72. 2024. View Article : Google Scholar : PubMed/NCBI

10 

Fantone S, Tossetta G, Cianfruglia L, Frontini A, Armeni T, Procopio AD, Pugnaloni A, Gualtieri AF and Marzioni D: Mechanisms of action of mineral fibres in a placental syncytiotrophoblast model: An in vitro toxicology study. Chem Biol Interact. 390:1108952024. View Article : Google Scholar : PubMed/NCBI

11 

Abrantes-Soares F, Lorigo M and Cairrao E: Effects of BPA substitutes on the prenatal and cardiovascular systems. Crit Rev Toxicol. 52:469–498. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Khan B, Allah Yar R, Khakwani AK, Karim S and Arslan Ali H: Preeclampsia incidence and its maternal and neonatal outcomes with associated risk factors. Cureus. 14:e311432022.PubMed/NCBI

13 

No authors listed: ACOG practice bulletin No. 202: Gestational hypertension and preeclampsia. Obstet Gynecol. 133:12019.

14 

Magee LA, Brown MA, Hall DR, Gupte S, Hennessy A, Karumanchi SA, Kenny LC, McCarthy F, Myers J, Poon LC, et al: The 2021 international society for the study of hypertension in pregnancy classification, diagnosis & management recommendations for international practice. Pregnancy Hypertens. 27:148–169. 2022. View Article : Google Scholar : PubMed/NCBI

15 

Chang KJ, Seow KM and Chen KH: Preeclampsia: Recent advances in predicting, preventing, and managing the maternal and fetal life-threatening condition. Int J Environ Res Public Health. 20:29942023. View Article : Google Scholar : PubMed/NCBI

16 

Gesuita R, Licini C, Picchiassi E, Tarquini F, Coata G, Fantone S, Tossetta G, Ciavattini A, Castellucci M, Di Renzo GC, et al: Association between first trimester plasma htra1 level and subsequent preeclampsia: A possible early marker? Pregnancy Hypertens. 18:58–62. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Deshpande JS, Sundrani DP, Sahay AS, Gupte SA and Joshi SR: Unravelling the potential of angiogenic factors for the early prediction of preeclampsia. Hypertens Res. 44:756–769. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Fantone S, Mazzucchelli R, Giannubilo SR, Ciavattini A, Marzioni D and Tossetta G: AT-rich interactive domain 1A protein expression in normal and pathological pregnancies complicated by preeclampsia. Histochem Cell Biol. 154:339–346. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Aneman I, Pienaar D, Suvakov S, Simic TP, Garovic VD and McClements L: Mechanisms of key innate immune cells in early- and late-onset preeclampsia. Front Immunol. 11:18642020. View Article : Google Scholar : PubMed/NCBI

20 

Marin R, Chiarello DI, Abad C, Rojas D, Toledo F and Sobrevia L: Oxidative stress and mitochondrial dysfunction in early-onset and late-onset preeclampsia. Biochim Biophys Acta Mol Basis Dis. 1866:1659612020. View Article : Google Scholar : PubMed/NCBI

21 

Staff AC: The two-stage placental model of preeclampsia: An update. J Reprod Immunol. 134-135:1–10. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Örgül G, Aydın Haklı D, Özten G, Fadiloğlu E, Tanacan A and Beksaç MS: First trimester complete blood cell indices in early and late onset preeclampsia. Turk J Obstet Gynecol. 16:112–117. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Marzioni D, Lorenzi T, Altobelli E, Giannubilo SR, Paolinelli F, Tersigni C, Crescimanno C, Monsurrò V, Tranquilli AL, Di Simone N and Castellucci M: Alterations of maternal plasma HTRA1 level in preeclampsia complicated by IUGR. Placenta. 33:1036–1038. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Inversetti A, Pivato CA, Cristodoro M, Latini AC, Condorelli G, Di Simone N and Stefanini G: Update on long-term cardiovascular risk after pre-eclampsia: A systematic review and meta-analysis. Eur Heart J Qual Care Clin Outcomes. 10:4–13. 2024. View Article : Google Scholar

25 

Saito S, Shiozaki A, Nakashima A, Sakai M and Sasaki Y: The role of the immune system in preeclampsia. Mol Aspects Med. 28:192–209. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Hahn S, Gupta AK, Troeger C, Rusterholz C and Holzgreve W: Disturbances in placental immunology: Ready for therapeutic interventions? Springer Semin Immunopathol. 27:477–493. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Boulanger H, Bounan S, Mahdhi A, Drouin D, Ahriz-Saksi S, Guimiot F and Rouas-Freiss N: Immunologic aspects of preeclampsia. AJOG Glob Rep. 4:1003212024. View Article : Google Scholar : PubMed/NCBI

28 

Conforti-Andreoni C, Spreafico R, Qian HL, Riteau N, Ryffel B, Ricciardi-Castagnoli P and Mortellaro A: Uric acid-driven Th17 differentiation requires inflammasome-derived IL-1 and IL-18. J Immunol. 187:5842–5850. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Saito S, Shiozaki A, Sasaki Y, Nakashima A, Shima T and Ito M: Regulatory T cells and regulatory natural killer (NK) cells play important roles in feto-maternal tolerance. Semin Immunopathol. 29:115–122. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Saito S, Nakashima A, Shima T and Ito M: Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am J Reprod Immunol. 63:601–610. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Figueiredo AS and Schumacher A: The T helper type 17/regulatory T cell paradigm in pregnancy. Immunology. 148:13–21. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Ribeiro VR, Romao-Veiga M, Romagnoli GG, Matias ML, Nunes PR, Borges VTM, Peracoli JC and Peracoli MTS: Association between cytokine profile and transcription factors produced by T-cell subsets in early- and late-onset pre-eclampsia. Immunology. 152:163–173. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Eghbal-Fard S, Yousefi M, Heydarlou H, Ahmadi M, Taghavi S, Movasaghpour A, Jadidi-Niaragh F, Yousefi B, Dolati S, Hojjat-Farsangi M, et al: The imbalance of Th17/Treg axis involved in the pathogenesis of preeclampsia. J Cell Physiol. 234:5106–5116. 2019. View Article : Google Scholar

34 

Saito S and Sakai M: Th1/Th2 balance in preeclampsia. J Reprod Immunol. 59:161–173. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Kluger MA, Luig M, Wegscheid C, Goerke B, Paust HJ, Brix SR, Yan I, Mittrücker HW, Hagl B, Renner ED, et al: Stat3 programs Th17-specific regulatory T cells to control GN. J Am Soc Nephrol. 25:1291–1302. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Lee JH, Kim TH, Oh SJ, Yoo JY, Akira S, Ku BJ, Lydon JP and Jeong JW: Signal transducer and activator of transcription-3 (Stat3) plays a critical role in implantation via progesterone receptor in uterus. FASEB J. 27:2553–2563. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Suman P, Malhotra SS and Gupta SK: LIF-STAT signaling and trophoblast biology. JAKSTAT. 2:e251552013.

38 

Fitzgerald JS, Busch S, Wengenmayer T, Foerster K, de la Motte T, Poehlmann TG and Markert UR: Signal transduction in trophoblast invasion. Chem Immunol Allergy. 88:181–199. 2005.PubMed/NCBI

39 

DiFederico E, Genbacev O and Fisher SJ: Preeclampsia is associated with widespread apoptosis of placental cytotrophoblasts within the uterine wall. Am J Pathol. 155:293–301. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Tossetta G, Fantone S, Giannubilo SR, Ciavattini A, Senzacqua M, Frontini A and Marzioni D: HTRA1 in placental cell models: A possible role in preeclampsia. Curr Issues Mol Biol. 45:3815–3828. 2023. View Article : Google Scholar : PubMed/NCBI

41 

He G, Xu W, Chen Y, Liu X and Xi M: Abnormal apoptosis of trophoblastic cells is related to the up-regulation of CYP11A gene in placenta of preeclampsia patients. PLoS One. 8:e596092013. View Article : Google Scholar : PubMed/NCBI

42 

Kisseleva T, Bhattacharya S, Braunstein J and Schindler CW: Signaling through the JAK/STAT pathway, recent advances and future challenges. Gene. 285:1–24. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Gupta S, Yan H, Wong LH, Ralph S, Krolewski J and Schindler C: The SH2 domains of Stat1 and Stat2 mediate multiple interactions in the transduction of IFN-alpha signals. EMBO J. 15:1075–1084. 1996. View Article : Google Scholar : PubMed/NCBI

44 

El-Tanani M, Al Khatib AO, Aladwan SM, Abuelhana A, McCarron PA and Tambuwala MM: Importance of STAT3 signaling in cancer, metastasis and therapeutic interventions. Cell Signal. 92:1102752022. View Article : Google Scholar

45 

Blake S, Hughes TP, Mayrhofer G and Lyons AB: The Src/ABL kinase inhibitor dasatinib (BMS-354825) inhibits function of normal human T-lymphocytes in vitro. Clin Immunol. 127:330–339. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Cheranov SY, Karpurapu M, Wang D, Zhang B, Venema RC and Rao GN: An essential role for SRC-activated STAT-3 in 14,15-EET-induced VEGF expression and angiogenesis. Blood. 111:5581–5591. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Murray PJ: The JAK-STAT signaling pathway: Input and output integration. J Immunol. 178:2623–2629. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Cortés-Ballinas L, López-Pérez TV and Rocha-Zavaleta L: STAT3 and the STAT3-regulated inhibitor of apoptosis protein survivin as potential therapeutic targets in colorectal cancer (Review). Biomed Rep. 21:1752024. View Article : Google Scholar

49 

Teng Y, Ross JL and Cowell JK: The involvement of JAK-STAT3 in cell motility, invasion, and metastasis. JAKSTAT. 3:e280862014.PubMed/NCBI

50 

Fang Y, Feng X, Xue N, Cao Y, Zhou P and Wei Z: STAT3 signaling pathway is involved in the pathogenesis of miscarriage. Placenta. 101:30–38. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Fitzgerald JS, Poehlmann TG, Schleussner E and Markert UR: Trophoblast invasion: The role of intracellular cytokine signaling via signal transducer and activator of transcription 3 (STAT3). Hum Reprod Update. 14:335–344. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Borg AJ, Yong HEJ, Lappas M, Degrelle SA, Keogh RJ, Da Silva-Costa F, Fournier T, Abumaree M, Keelan JA, Kalionis B and Murthi P: Decreased STAT3 in human idiopathic fetal growth restriction contributes to trophoblast dysfunction. Reproduction. 149:523–532. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Weber M, Kuhn C, Schulz S, Schiessl B, Schleussner E, Jeschke U, Markert UR and Fitzgerald JS: Expression of signal transducer and activator of transcription 3 (STAT3) and its activated forms is negatively altered in trophoblast and decidual stroma cells derived from preeclampsia placentae. Histopathology. 60:657–662. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Travaglino A, Raffone A, Saccone G, Migliorini S, Maruotti GM, Esposito G, Mollo A, Martinelli P, Zullo F and D'Armiento M: Placental morphology, apoptosis, angiogenesis and epithelial mechanisms in early-onset preeclampsia. Eur J Obstet Gynecol Reprod Biol. 234:200–206. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Zhang Z, Yang X, Zhang L, Duan Z, Jia L, Wang P, Shi Y, Li Y and Gao J: Decreased expression and activation of Stat3 in severe preeclampsia. J Mol Histol. 46:205–219. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Yang HY: MiR-133b regulates oxidative stress injury of trophoblasts in preeclampsia by mediating the JAK2/STAT3 signaling pathway. J Mol Histol. 52:1177–1188. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Zhou W, Wang H, Yang J, Long W, Zhang B, Liu J and Yu B: Down-regulated circPAPPA suppresses the proliferation and invasion of trophoblast cells via the miR-384/STAT3 pathway. Biosci Rep. 39:BSR201919652019. View Article : Google Scholar : PubMed/NCBI

58 

Liu M, Liao L, Gao Y, Yin Y, Wei X, Xu Q, Gao L and Zhou R: BCAM deficiency may contribute to preeclampsia by suppressing the PIK3R6/p-STAT3 signaling. Hypertension. 79:2830–2842. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Xu X, Zhu M, Zu Y, Wang G, Li X and Yan J: Nox2 inhibition reduces trophoblast ferroptosis in preeclampsia via the STAT3/GPX4 pathway. Life Sci. 343:1225552024. View Article : Google Scholar : PubMed/NCBI

60 

Qu HM, Qu LP, Li XY and Pan XZ: Overexpressed HO-1 is associated with reduced STAT3 activation in preeclampsia placenta and inhibits STAT3 phosphorylation in placental JEG-3 cells under hypoxia. Arch Med Sci. 14:597–607. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Zhang Z, Wang X, Wang J and Zhang L: The decreased expression of Stat3 and p-Stat3 in preeclampsia-like rat placenta. J Mol Histol. 49:175–183. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Wang H, Liu ML, Chu C, Yu SJ, Li J, Shen HC, Meng Q and Zhang T: Paeonol alleviates placental inflammation and apoptosis in preeclampsia by inhibiting the JAK2/STAT3 signaling pathway. Kaohsiung J Med Sci. 38:1103–1112. 2022. View Article : Google Scholar : PubMed/NCBI

63 

Wang GJ, Yang Z, Huai J and Xiang QQ: Pravastatin alleviates oxidative stress and decreases placental trophoblastic cell apoptosis through IL-6/STAT3 signaling pathway in preeclampsia rats. Eur Rev Med Pharmacol Sci. 24:12955–12962. 2020.PubMed/NCBI

64 

Abdelzaher WY, Mostafa-Hedeab G, Bahaa HA, Mahran A, Atef Fawzy M, Abdel Hafez SMN, Welson NN and Rofaeil RR: Leukotriene receptor antagonist, montelukast ameliorates L-NAME-induced pre-eclampsia in rats through suppressing the IL-6/Jak2/STAT3 signaling pathway. Pharmaceuticals (Basel). 15:9142022. View Article : Google Scholar : PubMed/NCBI

65 

George EM and Arany I: Induction of heme oxygenase-1 shifts the balance from proinjury to prosurvival in the placentas of pregnant rats with reduced uterine perfusion pressure. Am J Physiol Regul Integr Comp Physiol. 302:R620–R626. 2012. View Article : Google Scholar : PubMed/NCBI

66 

Saad AF, Diken ZM, Kechichian TB, Clark SM, Olson GL, Saade GR and Costantine MM: Pravastatin effects on placental prosurvival molecular pathways in a mouse model of preeclampsia. Reprod Sci. 23:1593–1599. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Lecarpentier E and Tsatsaris V: Angiogenic balance (sFlt-1/PlGF) and preeclampsia. Ann Endocrinol (Paris). 77:97–100. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Kar M: Role of biomarkers in early detection of preeclampsia. J Clin Diagn Res. 8:BE01–BE04. 2014.PubMed/NCBI

69 

Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, Schisterman EF, Thadhani R, Sachs BP, Epstein FH, et al: Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 350:672–683. 2004. View Article : Google Scholar : PubMed/NCBI

70 

van Meeteren LA, Goumans MJ and ten Dijke P: TGF-β receptor signaling pathways in angiogenesis; emerging targets for anti-angiogenesis therapy. Curr Pharm Biotechnol. 12:2108–2120. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Ives CW, Sinkey R, Rajapreyar I, Tita ATN and Oparil S: Preeclampsia-pathophysiology and clinical presentations: JACC state-of-the-art review. J Am Coll Cardiol. 76:1690–1702. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Rajakumar A, Michael HM, Rajakumar PA, Shibata E, Hubel CA, Karumanchi SA, Thadhani R, Wolf M, Harger G and Markovic N: Extra-placental expression of vascular endothelial growth factor receptor-1, (Flt-1) and soluble Flt-1 (sFlt-1), by peripheral blood mononuclear cells (PBMCs) in normotensive and preeclamptic pregnant women. Placenta. 26:563–573. 2005. View Article : Google Scholar : PubMed/NCBI

73 

Zolfaghari MA, Motavalli R, Soltani-Zangbar MS, Parhizkar F, Danaii S, Aghebati-Maleki L, Noori M, Dolati S, Ahmadi M, Samadi Kafil H, et al: A new approach to the preeclampsia puzzle; MicroRNA-326 in CD4+ lymphocytes might be as a potential suspect. J Reprod Immunol. 145:1033172021. View Article : Google Scholar

74 

Mathur AN, Chang HC, Zisoulis DG, Stritesky GL, Yu Q, O'Malley JT, Kapur R, Levy DE, Kansas GS and Kaplan MH: Stat3 and Stat4 direct development of IL-17-secreting Th cells. J Immunol. 178:4901–4907. 2007. View Article : Google Scholar : PubMed/NCBI

75 

Davis GK, Fehrenbach DJ and Madhur MS: Interleukin 17A: Key player in the pathogenesis of hypertension and a potential therapeutic target. Curr Hypertens Rep. 23:132021. View Article : Google Scholar : PubMed/NCBI

76 

Bowman T, Garcia R, Turkson J and Jove R: STATs in oncogenesis. Oncogene. 19:2474–2488. 2000. View Article : Google Scholar : PubMed/NCBI

77 

Bushway ME, Gerber SA, Fenton BM, Miller RK, Lord EM and Murphy SP: Morphological and phenotypic analyses of the human placenta using whole mount immunofluorescence. Biol Reprod. 90:1102014. View Article : Google Scholar : PubMed/NCBI

78 

Zozzaro-Smith PE, Bushway ME, Gerber SA, Hebert D, Pressman EK, Lord EM, Miller RK and Murphy SP: Whole mount immunofluorescence analysis of placentas from normotensive versus preeclamptic pregnancies. Placenta. 36:1310–1317. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Lykke JA, Langhoff-Roos J, Sibai BM, Funai EF, Triche EW and Paidas MJ: Hypertensive pregnancy disorders and subsequent cardiovascular morbidity and type 2 diabetes mellitus in the mother. Hypertension. 53:944–951. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Männistö T, Mendola P, Vääräsmäki M, Järvelin MR, Hartikainen AL, Pouta A and Suvanto E: Elevated blood pressure in pregnancy and subsequent chronic disease risk. Circulation. 127:681–690. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Rana S, Lemoine E, Granger JP and Karumanchi SA: Preeclampsia: Pathophysiology, challenges, and perspectives. Circ Res. 124:1094–1112. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Christensen M, Petersen JL, Sivanandam P, Kronborg CS, Knudsen UB and Martensen PM: Reduction of serum-induced endothelial STAT3(Y705) activation is associated with preeclampsia. Pregnancy Hypertens. 25:103–109. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Harmon AC, Cornelius DC, Amaral LM, Faulkner JL, Cunningham MW Jr, Wallace K and LaMarca B: The role of inflammation in the pathology of preeclampsia. Clin Sci (Lond). 130:409–419. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Tossetta G, Fantone S, Licini C, Marzioni D and Mattioli-Belmonte M: The multifaced role of HtrA1 in the development of joint and skeletal disorders. Bone. 157:1163502022. View Article : Google Scholar : PubMed/NCBI

85 

Tossetta G, Fantone S, Busilacchi EM, Di Simone N, Giannubilo SR, Scambia G, Giordano A and Marzioni D: Modulation of matrix metalloproteases by ciliary neurotrophic factor in human placental development. Cell Tissue Res. 390:113–129. 2022. View Article : Google Scholar : PubMed/NCBI

86 

Tossetta G, Fantone S, Piani F, Crescimanno C, Ciavattini A, Giannubilo SR and Marzioni D: Modulation of NRF2/KEAP1 signaling in preeclampsia. Cells. 12:15452023. View Article : Google Scholar : PubMed/NCBI

87 

Tossetta G, Fantone S, Marzioni D and Mazzucchelli R: Role of natural and synthetic compounds in modulating NRF2/KEAP1 signaling pathway in prostate cancer. Cancers (Basel). 15:30372023. View Article : Google Scholar : PubMed/NCBI

88 

Ray PP, Islam MA, Islam MS, Han A, Geng P, Aziz MA and Mamun AA: A comprehensive evaluation of the therapeutic potential of silibinin: A ray of hope in cancer treatment. Front Pharmacol. 15:13497452024. View Article : Google Scholar : PubMed/NCBI

89 

Xu LN, Xu RA, Zhang D, Su SS, Xu HY, Wu Q and Li YP: The changes of expressive levels of IL-17A, STAT3, and RORγt in different invasive pulmonary aspergillosis mice. Infect Drug Resist. 11:1321–1328. 2018. View Article : Google Scholar :

90 

Ribeiro VR, Romao-Veiga M, Nunes PR, de Oliveira LRC, Romagnoli GG, Peracoli JC and Peracoli MTS: Silibinin downregulates the expression of the Th1 and Th17 profiles by modulation of STATs and transcription factors in pregnant women with preeclampsia. Int Immunopharmacol. 109:1088072022. View Article : Google Scholar : PubMed/NCBI

91 

Zhang L, Li DC and Liu LF: Paeonol: Pharmacological effects and mechanisms of action. Int Immunopharmacol. 72:413–421. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Chen X, Zhang Z, Zhang X, Jia Z, Liu J, Chen X, Xu A, Liang X and Li G: Paeonol attenuates heart failure induced by transverse aortic constriction via ERK1/2 signalling. Pharm Biol. 60:562–569. 2022. View Article : Google Scholar : PubMed/NCBI

93 

Feldman D, Krishnan AV, Swami S, Giovannucci E and Feldman BJ: The role of vitamin D in reducing cancer risk and progression. Nat Rev Cancer. 14:342–357. 2014. View Article : Google Scholar : PubMed/NCBI

94 

Zhang H, Shih DQ and Zhang X: Mechanisms underlying effects of 1,25-Dihydroxyvitamin D3 on the Th17 cells. Eur J Microbiol Immunol (Bp). 3:237–240. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Sassi F, Tamone C and D'Amelio P: Vitamin D: Nutrient, hormone, and immunomodulator. Nutrients. 10:16562018. View Article : Google Scholar : PubMed/NCBI

96 

Xu L, Lee M, Jeyabalan A and Roberts JM: The relationship of hypovitaminosis D and IL-6 in preeclampsia. Am J Obstet Gynecol. 210:149.e1–e7. 2014. View Article : Google Scholar

97 

Ribeiro VR, Romao-Veiga M, Nunes PR, de Oliveira LRC, Romagnoli GG, Peracoli JC and Peracoli MTS: Immunomodulatory effect of vitamin D on the STATs and transcription factors of CD4+ T cell subsets in pregnant women with preeclampsia. Clin Immunol. 234:1089172022. View Article : Google Scholar

98 

Bhasin D, Cisek K, Pandharkar T, Regan N, Li C, Pandit B, Lin J and Li PK: Design, synthesis, and studies of small molecule STAT3 inhibitors. Bioorg Med Chem Lett. 18:391–395. 2008. View Article : Google Scholar

99 

Huang W, Dong Z, Wang F, Peng H, Liu JY and Zhang JT: A small molecule compound targeting STAT3 DNA-binding domain inhibits cancer cell proliferation, migration, and invasion. ACS Chem Biol. 9:1188–1196. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Schust J, Sperl B, Hollis A, Mayer TU and Berg T: Stattic: A small-molecule inhibitor of STAT3 activation and dimerization. Chem Biol. 13:1235–1242. 2006. View Article : Google Scholar : PubMed/NCBI

101 

Mendola P, Nobles C, Williams A, Sherman S, Kanner J, Seeni I and Grantz K: Air pollution and preterm birth: Do air pollution changes over time influence risk in consecutive pregnancies among low-risk women? Int J Environ Res Public Health. 16:33652019. View Article : Google Scholar : PubMed/NCBI

102 

Yang S, Tan Y, Mei H, Wang F, Li N, Zhao J, Zhang Y, Qian Z, Chang JJ, Syberg KM, et al: Ambient air pollution the risk of stillbirth: A prospective birth cohort study in Wuhan, China. Int J Hyg Environ Health. 221:502–509. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Zhang H, Dong H, Ren M, Liang Q, Shen X, Wang Q, Yu L, Lin H, Luo Q, Chen W, et al: Ambient air pollution exposure and gestational diabetes mellitus in Guangzhou, China: A prospective cohort study. Sci Total Environ. 699:1343902020. View Article : Google Scholar

104 

Wang Q, Zhang H, Liang Q, Knibbs LD, Ren M, Li C, Bao J, Wang S, He Y, Zhu L, et al: Effects of prenatal exposure to air pollution on preeclampsia in Shenzhen, China. Environ Pollut. 237:18–27. 2018. View Article : Google Scholar : PubMed/NCBI

105 

Hu L, Huang B, Bai S, Tan J, Liu Y and Chen H, Liu Y, Zhu L, Zhang J and Chen H: SO2 derivatives induce dysfunction in human trophoblasts via inhibiting ROS/IL-6/STAT3 pathway. Ecotoxicol Environ Saf. 210:1118722021. View Article : Google Scholar

106 

Sirtori CR: The pharmacology of statins. Pharmacol Res. 88:3–11. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Marrs CC and Costantine MM: Should we add pravastatin to aspirin for preeclampsia prevention in high-risk women? Clin Obstet Gynecol. 60:161–168. 2017. View Article : Google Scholar :

108 

Lu F, Bytautiene E, Tamayo E, Gamble P, Anderson GD, Hankins GD, Longo M and Saade GR: Gender-specific effect of overexpression of sFlt-1 in pregnant mice on fetal programming of blood pressure in the offspring later in life. Am J Obstet Gynecol. 197:418.e1–e5. 2007. View Article : Google Scholar : PubMed/NCBI

109 

Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, Libermann TA, Morgan JP, Sellke FW, Stillman IE, et al: Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest. 111:649–658. 2003. View Article : Google Scholar : PubMed/NCBI

110 

Korthals-de Bos I, Van Tulder M, Boers M, Verhoeven AC, Adèr HJ, Bibo J, Boonen A and Van Der Linden S: Indirect and total costs of early rheumatoid arthritis: A randomized comparison of combined step-down prednisolone, methotrexate, and sulfasalazine with sulfasalazine alone. J Rheumatol. 31:1709–1716. 2004.PubMed/NCBI

111 

Cai C, Lu J, Lai L, Song D, Shen J, Tong J, Zheng Q, Wu K, Qian J and Ran Z: Drug therapy and monitoring for inflammatory bowel disease: A multinational questionnaire investigation in Asia. Intest Res. 20:213–223. 2022. View Article : Google Scholar : PubMed/NCBI

112 

Hastie R, Brownfoot FC, Pritchard N, Hannan NJ, Cannon P, Nguyen V, Palmer K, Beard S, Tong S and Kaitu'u-Lino TJ: EGFR (epidermal growth factor receptor) signaling and the mitochondria regulate sFlt-1 (soluble FMS-like tyrosine kinase-1) secretion. Hypertension. 73:659–670. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Hastie R, Brownfoot FC, Cannon P, Nguyen V, Tuohey L, Hannan NJ, Tong S and Kaitu'u-Lino TJ: Sulfasalazine decreases soluble fms-like tyrosine kinase-1 secretion potentially via inhibition of upstream placental epidermal growth factor receptor signaling. Placenta. 87:53–57. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Fantel AG, Nekahi N, Shepard TH, Cornel LM, Unis AS and Lemire RJ: The teratogenicity of N(omega)-nitro-L-ariginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, in rats. Reprod Toxicol. 11:709–717. 1997. View Article : Google Scholar : PubMed/NCBI

115 

Hao H, Li F, Wang F, Ran J, Chen Y, Yang L, Ma H, Wang J and Yang H: Protective effect of metformin on the NG-nitro-l-arginine methyl ester (l-NAME)-induced rat models of preeclampsia. Biochem Biophys Res Commun. 739:1509962024. View Article : Google Scholar : PubMed/NCBI

116 

Liu S, Gao M, Zhang X, Wei J and Cui H: FOXP2 overexpression upregulates LAMA4 expression and thereby alleviates preeclampsia by regulating trophoblast behavior. Commun Biol. 7:14272024. View Article : Google Scholar : PubMed/NCBI

117 

Fitriana F, Soetrisno S, Sulistyowati S and Indarto D: Evaluation of placental bed uterine in L-NAME-induced early-onset preeclampsia (EO-PE) like the rat model. Turk J Obstet Gynecol. 21:180–189. 2024. View Article : Google Scholar : PubMed/NCBI

118 

Tomruk C, Şirin Tomruk C, Denizlioğlu B, Olukman M, Ercan G, Duman S, Köse T, Çetin Uyanıkgil EÖ, Uyanıkgil Y and Uysal A: Effects of apelin on neonatal brain neurogenesis in L-NAME-induced maternal preeclampsia. Sci Rep. 14:193472024. View Article : Google Scholar : PubMed/NCBI

119 

Palmsten K, Flores KF, Chambers CD, Weiss LA, Sundaram R and Buck Louis GM: Most frequently reported prescription medications and supplements in couples planning pregnancy: The LIFE study. Reprod Sci. 25:94–101. 2018. View Article : Google Scholar :

120 

Walia M, Lodha R and Kabra SK: Montelukast in pediatric asthma management. Indian J Pediatr. 73:275–282. 2006. View Article : Google Scholar : PubMed/NCBI

121 

Yerraguravagari B, Penchikala NP, Kolusu AS, Ganesh GS, Konduri P, Nemmani KVS and Samudrala PK: Montelukast ameliorates scopolamine-induced Alzheimer's disease: Role on cholinergic neurotransmission, antioxidant defence system, neuroinflammation and expression of BDNF. CNS Neurol Disord Drug Targets. 23:1040–1055. 2024. View Article : Google Scholar

122 

Li P, Ma X, Huang D and Gu X: Exploring the roles of non-coding RNAs in liver regeneration. Noncoding RNA Res. 9:945–953. 2024. View Article : Google Scholar : PubMed/NCBI

123 

Hill M and Tran N: miRNA interplay: Mechanisms and consequences in cancer. Dis Model Mech. 14:dmm0476622021. View Article : Google Scholar : PubMed/NCBI

124 

Dong K, Hou Y, Zhang N, Duan B, Ma A and Zhang Z: Down-regulated placental miR-21 contributes to preeclampsia through targeting RASA1. Hypertens Pregnancy. 40:236–245. 2021. View Article : Google Scholar : PubMed/NCBI

125 

He H, Zhang H, Li Z, Wang R, Li N and Zhu L: miRNA-214: Expression, therapeutic and diagnostic potential in cancer. Tumori. 101:375–383. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Li D, Xia L, Chen M, Lin C, Wu H, Zhang Y, Pan S and Li X: miR-133b, a particular member of myomiRs, coming into playing its unique pathological role in human cancer. Oncotarget. 8:50193–50208. 2017. View Article : Google Scholar : PubMed/NCBI

127 

Wang X, Li B, Wang J, Lei J, Liu C, Ma Y and Zhao H: Evidence that miR-133a causes recurrent spontaneous abortion by reducing HLA-G expression. Reprod Biomed Online. 25:415–424. 2012. View Article : Google Scholar : PubMed/NCBI

128 

Licini C, Avellini C, Picchiassi E, Mensà E, Fantone S, Ramini D, Tersigni C, Tossetta G, Castellucci C, Tarquini F, et al: Pre-eclampsia predictive ability of maternal miR-125b: A clinical and experimental study. Transl Res. 228:13–27. 2021. View Article : Google Scholar

129 

Tang J, Wang D, Lu J and Zhou X: MiR-125b participates in the occurrence of preeclampsia by regulating the migration and invasion of extravillous trophoblastic cells through STAT3 signaling pathway. J Recept Signal Transduct Res. 41:202–208. 2021. View Article : Google Scholar

130 

Song X, Luo X, Gao Q, Wang Y, Gao Q and Long W: Dysregulation of LncRNAs in placenta and pathogenesis of preeclampsia. Curr Drug Targets. 18:1165–1170. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Zhang TN, Wang W, Huang XM and Gao SY: Non-coding RNAs and extracellular vehicles: Their role in the pathogenesis of gestational diabetes mellitus. Front Endocrinol (Lausanne). 12:6642872021. View Article : Google Scholar : PubMed/NCBI

132 

Wang P, Xue Y, Han Y, Lin L, Wu C, Xu S, Jiang Z, Xu J, Liu Q and Cao X: The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science. 344:310–313. 2014. View Article : Google Scholar : PubMed/NCBI

133 

Faas MM and De Vos P: Innate immune cells in the placental bed in healthy pregnancy and preeclampsia. Placenta. 69:125–133. 2018. View Article : Google Scholar : PubMed/NCBI

134 

Zhang W, Zhou Y and Ding Y: Lnc-DC mediates the over-maturation of decidual dendritic cells and induces the increase in Th1 cells in preeclampsia. Am J Reprod Immunol. 77:e126472017. View Article : Google Scholar

135 

Zhang W, Yang M, Yu L, Hu Y, Deng Y, Liu Y, Xiao S and Ding Y: Long non-coding RNA lnc-DC in dendritic cells regulates trophoblast invasion via p-STAT3-mediated TIMP/MMP expression. Am J Reprod Immunol. 83:e132392020. View Article : Google Scholar : PubMed/NCBI

136 

Gao Q, Wang T, Pan L, Qian C, Wang J, Xin Q, Liu Y, Zhang Z, Xu Y, He X and Cao Y: Circular RNAs: Novel potential regulators in embryogenesis, female infertility, and pregnancy-related diseases. J Cell Physiol. 236:7223–7241. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Zhou L, Duan Y, Fu K, Zhang M, Li K and Yin R: The role of B7-H4 in ovarian cancer immunotherapy: Current status, challenges, and perspectives. Front Immunol. 15:14260502024. View Article : Google Scholar : PubMed/NCBI

138 

Duan L, Reisch B, Iannaccone A, Hadrovic E, Wu Y, Vogtmann R, Winterhager E, Kimmig R, Köninger A, Mach P and Gellhaus A: Abnormal expression of the costimulatory molecule B7-H4 in placental chorionic villous and decidual basalis tissues of patients with preeclampsia and HELLP syndrome. Am J Reprod Immunol. 86:e134302021. View Article : Google Scholar : PubMed/NCBI

139 

Mach P, Nolte-Boenigk L, Droste L, Fox L, Frank M, Schmidt B, Herse F, Verlohren S, Wicherek L, Iannaccone A, et al: Soluble B7-H4 blood serum levels are elevated in women at high risk for preeclampsia in the first trimester, as well as in patients with confirmed preeclampsia. Am J Reprod Immunol. 80:e129882018. View Article : Google Scholar : PubMed/NCBI

140 

Ma Y, Duan L, Reisch B, Kimmig R, Iannaccone A and Gellhaus A: Impact of the immunomodulatory factor soluble B7-H4 in the progress of preeclampsia by inhibiting essential functions of extravillous trophoblast cells. Cells. 13:13722024. View Article : Google Scholar : PubMed/NCBI

141 

Li H, Zhou L and Dai J: Retinoic acid receptor-related orphan receptor RORα regulates differentiation and survival of keratinocytes during hypoxia. J Cell Physiol. 233:641–650. 2018. View Article : Google Scholar

142 

Chauvet C, Bois-Joyeux B and Danan JL: Retinoic acid receptor-related orphan receptor (ROR) alpha4 is the predominant isoform of the nuclear receptor RORalpha in the liver and is up-regulated by hypoxia in HepG2 human hepatoma cells. Biochem J. 364:449–456. 2002. View Article : Google Scholar : PubMed/NCBI

143 

Yu Y and Zhu T: RAR-related orphan receptor: An accelerated preeclampsia progression by activating the JAK/STAT3 pathway. Yonsei Med J. 63:554–563. 2022. View Article : Google Scholar : PubMed/NCBI

144 

Kikkawa Y, Ogawa T, Sudo R, Yamada Y, Katagiri F, Hozumi K, Nomizu M and Miner JH: The lutheran/basal cell adhesion molecule promotes tumor cell migration by modulating integrin-mediated cell attachment to laminin-511 protein. J Biol Chem. 288:30990–31001. 2013. View Article : Google Scholar : PubMed/NCBI

145 

Yang WH, Huang Z, Wu J, Ding CC, Murphy SK and Chi JT: A TAZ-ANGPTL4-NOX2 axis regulates ferroptotic cell death and chemoresistance in epithelial ovarian cancer. Mol Cancer Res. 18:79–90. 2020. View Article : Google Scholar :

146 

Cao Y, Luo F, Peng J, Fang Z, Liu Q and Zhou S: KMT2B-dependent RFK transcription activates the TNF-α/NOX2 pathway and enhances ferroptosis caused by myocardial ischemia-reperfusion. J Mol Cell Cardiol. 173:75–91. 2022. View Article : Google Scholar : PubMed/NCBI

147 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

148 

Zhao Y, Wang C, Yang T, Wang H, Zhao S, Sun N, Chen Y, Zhang H and Fan H: Chlorogenic Acid alleviates chronic stress-induced duodenal ferroptosis via the inhibition of the IL-6/JAK2/STAT3 signaling pathway in rats. J Agric Food Chem. 70:4353–4361. 2022. View Article : Google Scholar : PubMed/NCBI

149 

Zhang W, Gong M, Zhang W, Mo J, Zhang S, Zhu Z, Wang X, Zhang B, Qian W, Wu Z, et al: Thiostrepton induces ferroptosis in pancreatic cancer cells through STAT3/GPX4 signaling. Cell Death Dis. 13:6302022. View Article : Google Scholar

150 

Jiang Z, Li S, Han MJ, Hu GM and Cheng P: High expression of NSUN5 promotes cell proliferation via cell cycle regulation in colorectal cancer. Am J Transl Res. 12:3858–3870. 2020.PubMed/NCBI

151 

Zhang H, Li H, Yao J, Zhao M and Zhang C: The mutation of NSUN5 R295C promotes preeclampsia by impairing decidualization through downregulating IL-11Rα. iScience. 27:1088992024. View Article : Google Scholar

152 

Graham AM and Presnell JS: Hypoxia inducible factor (HIF) transcription factor family expansion, diversification, divergence and selection in eukaryotes. PLoS One. 12:e01795452017. View Article : Google Scholar : PubMed/NCBI

153 

Qu H, Yu Q, Jia B, Zhou W, Zhang Y and Mu L: HIF-3α affects preeclampsia development by regulating EVT growth via activation of the Flt-1/JAK/STAT signaling pathway in hypoxia. Mol Med Rep. 23:682021. View Article : Google Scholar

154 

Gerke V and Moss SE: Annexins: From structure to function. Physiol Rev. 82:331–371. 2002. View Article : Google Scholar : PubMed/NCBI

155 

Alli-Shaik A, Wee S, Lim LHK and Gunaratne J: Phosphoproteomics reveals network rewiring to a pro-adhesion state in annexin-1-deficient mammary epithelial cells. Breast Cancer Res. 19:1322017. View Article : Google Scholar : PubMed/NCBI

156 

Lee SH, Lee PH, Kim BG, Seo HJ, Baek AR, Park JS, Lee JH, Park SW, Kim DJ, Park CS and Jang AS: Annexin A1 in plasma from patients with bronchial asthma: Its association with lung function. BMC Pulm Med. 18:12018. View Article : Google Scholar : PubMed/NCBI

157 

Perucci LO, Carneiro FS, Ferreira CN, Sugimoto MA, Soriani FM, Martins GG, Lima KM, Guimarães FL, Teixeira AL, Dusse LM, et al: Annexin A1 is increased in the plasma of preeclamptic women. PLoS One. 10:e01384752015. View Article : Google Scholar : PubMed/NCBI

158 

Perucci LO, Vieira ÉLM, Teixeira AL, Gomes KB, Dusse LM and Sousa LP: Decreased plasma concentrations of brain-derived neurotrophic factor in preeclampsia. Clin Chim Acta. 464:142–147. 2017. View Article : Google Scholar

159 

Feng J, Wang X, Li H, Wang L and Tang Z: Silencing of Annexin A1 suppressed the apoptosis and inflammatory response of preeclampsia rat trophoblasts. Int J Mol Med. 42:3125–3134. 2018.PubMed/NCBI

160 

Xue Y, Luo M, Hu X, Li X, Shen J, Zhu W, Huang L, Hu Y, Guo Y, Liu L, et al: Macrophages regulate vascular smooth muscle cell function during atherosclerosis progression through IL-1β/STAT3 signaling. Commun Biol. 5:13162022. View Article : Google Scholar

161 

Cruceriu D, Baldasici O, Balacescu O and Berindan-Neagoe I: The dual role of tumor necrosis factor-alpha (TNF-α) in breast cancer: Molecular insights and therapeutic approaches. Cell Oncol (Dordr). 43:1–18. 2020. View Article : Google Scholar : PubMed/NCBI

162 

Sreenivasan L, Wang H, Yap SQ, Leclair P, Tam A and Lim CJ: Autocrine IL-6/STAT3 signaling aids development of acquired drug resistance in group 3 medulloblastoma. Cell Death Dis. 11:10352020. View Article : Google Scholar : PubMed/NCBI

163 

Ma JH, Qin L and Li X: Role of STAT3 signaling pathway in breast cancer. Cell Commun Signal. 18:332020. View Article : Google Scholar : PubMed/NCBI

164 

Mo HQ, Tian FJ, Li X, Zhang J, Ma XL, Zeng WH, Lin Y and Zhang Y: ANXA7 regulates trophoblast proliferation and apoptosis in preeclampsia. Am J Reprod Immunol. 82:e131832019. View Article : Google Scholar : PubMed/NCBI

165 

Xu C, Li X, Guo P and Wang J: Hypoxia-induced activation of JAK/STAT3 signaling pathway promotes trophoblast cell viability and angiogenesis in preeclampsia. Med Sci Monit. 23:4909–4917. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Yoshida H and Hunter CA: The immunobiology of interleukin-27. Annu Rev Immunol. 33:417–443. 2015. View Article : Google Scholar : PubMed/NCBI

167 

Yin N, Zhang H, Luo X, Ding Y, Xiao X, Liu X, Shan N, Zhang X, Deng Q, Zhuang B and Qi H: IL-27 activates human trophoblasts to express IP-10 and IL-6: implications in the immunopathophysiology of preeclampsia. Mediators Inflamm. 2014:9268752014. View Article : Google Scholar : PubMed/NCBI

168 

Ge H, Yin N, Han TL, Huang D, Chen X, Xu P, He C, Tong C and Qi H: Interleukin-27 inhibits trophoblast cell invasion and migration by affecting the epithelial-mesenchymal transition in preeclampsia. Reprod Sci. 26:928–938. 2019. View Article : Google Scholar

169 

Cao J, Inoue K, Li X, Drummond G and Abraham NG: Physiological significance of heme oxygenase in hypertension. Int J Biochem Cell Biol. 41:1025–1033. 2009. View Article : Google Scholar :

170 

Tsuchihashi S, Zhai Y, Bo Q, Busuttil RW and Kupiec-Weglinski JW: Heme oxygenase-1 mediated cytoprotection against liver ischemia and reperfusion injury: Inhibition of type-1 interferon signaling. Transplantation. 83:1628–1634. 2007. View Article : Google Scholar : PubMed/NCBI

171 

Csongradi E, Docarmo JM, Dubinion JH, Vera T and Stec DE: Chronic HO-1 induction with cobalt protoporphyrin (CoPP) treatment increases oxygen consumption, activity, heat production and lowers body weight in obese melanocortin-4 receptor-deficient mice. Int J Obes (Lond). 36:244–253. 2012. View Article : Google Scholar

172 

Downey JM, Davis AM and Cohen MV: Signaling pathways in ischemic preconditioning. Heart Fail Rev. 12:181–188. 2007. View Article : Google Scholar : PubMed/NCBI

173 

Xuan YT, Guo Y, Han H, Zhu Y and Bolli R: An essential role of the JAK-STAT pathway in ischemic preconditioning. Proc Natl Acad Sci USA. 98:9050–9055. 2001. View Article : Google Scholar : PubMed/NCBI

174 

Hilfiker-Kleiner D, Hilfiker A and Drexler H: Many good reasons to have STAT3 in the heart. Pharmacol Ther. 107:131–137. 2005. View Article : Google Scholar : PubMed/NCBI

175 

Andrés O, Kellermann T, López-Giráldez F, Rozas J, Domingo-Roura X and Bosch M: RPS4Y gene family evolution in primates. BMC Evol Biol. 8:1422008. View Article : Google Scholar : PubMed/NCBI

176 

Chen Y, Chen Y, Tang C, Zhao Q, Xu T, Kang Q, Jiang B and Zhang L: RPS4Y1 promotes high glucose-induced endothelial cell apoptosis and inflammation by activation of the p38 MAPK signaling. Diabetes Metab Syndr Obes. 14:4523–4534. 2021. View Article : Google Scholar : PubMed/NCBI

177 

Chen X, Tong C, Li H, Peng W, Li R, Luo X, Ge H, Ran Y, Li Q, Liu Y, et al: Dysregulated expression of RPS4Y1 (ribosomal protein S4, Y-linked 1) impairs STAT3 (signal transducer and activator of transcription 3) signaling to suppress trophoblast cell migration and invasion in preeclampsia. Hypertension. 71:481–490. 2018. View Article : Google Scholar : PubMed/NCBI

178 

Boonstra J, Rijken P, Humbel B, Cremers F, Verkleij A and van Bergen en Henegouwen P: The epidermal growth factor. Cell Biol Int. 19:413–430. 1995. View Article : Google Scholar : PubMed/NCBI

179 

Armant DR, Fritz R, Kilburn BA, Kim YM, Nien JK, Maihle NJ, Romero R and Leach RE: Reduced expression of the epidermal growth factor signaling system in preeclampsia. Placenta. 36:270–278. 2015. View Article : Google Scholar : PubMed/NCBI

180 

Malik A, Pal R and Gupta SK: Interdependence of JAK-STAT and MAPK signaling pathways during EGF-mediated HTR-8/SVneo cell invasion. PLoS One. 12:e01782692017. View Article : Google Scholar : PubMed/NCBI

181 

Chen LJ, Han ZQ, Zhou H, Zou L and Zou P: Inhibition of HLA-G expression via RNAi abolishes resistance of extravillous trophoblast cell line TEV-1 to NK lysis. Placenta. 31:519–527. 2010. View Article : Google Scholar : PubMed/NCBI

182 

Cecati M, Giannubilo SR, Emanuelli M, Tranquilli AL and Saccucci F: HLA-G and pregnancy adverse outcomes. Med Hypotheses. 76:782–784. 2011. View Article : Google Scholar : PubMed/NCBI

183 

Liu X, Gu W and Li X: HLA-G regulates the invasive properties of JEG-3 choriocarcinoma cells by controlling STAT3 activation. Placenta. 34:1044–1052. 2013. View Article : Google Scholar : PubMed/NCBI

184 

Huppertz B: The anatomy of the normal placenta. J Clin Pathol. 61:1296–1302. 2008. View Article : Google Scholar : PubMed/NCBI

185 

Kannaiyan J, Muthukutty P, Iqbal MDT and Paulraj B: Villous chorion: A potential source for pluripotent-like stromal cells. J Nat Sci Biol Med. 8:221–228. 2017. View Article : Google Scholar : PubMed/NCBI

186 

Chu Y, Zhu C, Yue C, Peng W, Chen W, He G, Liu C, Lv Y, Gao G, Yao K, et al: Chorionic villus-derived mesenchymal stem cell-mediated autophagy promotes the proliferation and invasiveness of trophoblasts under hypoxia by activating the JAK2/STAT3 signaling pathway. Cell Biosci. 11:1822021. View Article : Google Scholar

187 

Valesini G and Alessandri C: New facet of antiphospholipid antibodies. Ann N Y Acad Sci. 1051:487–497. 2005. View Article : Google Scholar : PubMed/NCBI

188 

Di Simone N, Caliandro D, Castellani R, Ferrazzani S and Caruso A: Interleukin-3 and human trophoblast: In vitro explanations for the effect of interleukin in patients with antiphospholipid antibody syndrome. Fertil Steril. 73:1194–1200. 2000. View Article : Google Scholar : PubMed/NCBI

189 

Branch DW and Khamashta MA: Antiphospholipid syndrome: Obstetric diagnosis, management, and controversies. Obstet Gynecol. 101:1333–1344. 2003.PubMed/NCBI

190 

Mulla MJ, Brosens JJ, Chamley LW, Giles I, Pericleous C, Rahman A, Joyce SK, Panda B, Paidas MJ and Abrahams VM: Antiphospholipid antibodies induce a pro-inflammatory response in first trimester trophoblast via the TLR4/MyD88 pathway. Am J Reprod Immunol. 62:96–111. 2009. View Article : Google Scholar : PubMed/NCBI

191 

Mulla MJ, Myrtolli K, Brosens JJ, Chamley LW, Kwak-Kim JY, Paidas MJ and Abrahams VM: Antiphospholipid antibodies limit trophoblast migration by reducing IL-6 production and STAT3 activity. Am J Reprod Immunol. 63:339–348. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2025
Volume 55 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Marzioni D, Piani F, Di Simone N, Giannubilo SR, Ciavattini A and Tossetta G: Importance of STAT3 signaling in preeclampsia (Review). Int J Mol Med 55: 58, 2025.
APA
Marzioni, D., Piani, F., Di Simone, N., Giannubilo, S.R., Ciavattini, A., & Tossetta, G. (2025). Importance of STAT3 signaling in preeclampsia (Review). International Journal of Molecular Medicine, 55, 58. https://doi.org/10.3892/ijmm.2025.5499
MLA
Marzioni, D., Piani, F., Di Simone, N., Giannubilo, S. R., Ciavattini, A., Tossetta, G."Importance of STAT3 signaling in preeclampsia (Review)". International Journal of Molecular Medicine 55.4 (2025): 58.
Chicago
Marzioni, D., Piani, F., Di Simone, N., Giannubilo, S. R., Ciavattini, A., Tossetta, G."Importance of STAT3 signaling in preeclampsia (Review)". International Journal of Molecular Medicine 55, no. 4 (2025): 58. https://doi.org/10.3892/ijmm.2025.5499