Open access peer-reviewed chapter

Perspective Chapter: Role of Cytotrophoblast Cells and Placenta-Derived Exosomes in Regulatory B Cell Differentiation and Function during Pregnancy

Written By

Gatien A.G. Lokossou and Maximin Senou

Submitted: 12 September 2022 Reviewed: 28 September 2022 Published: 18 January 2023

DOI: 10.5772/intechopen.108335

From the Edited Volume

Immunosuppression and Immunomodulation

Edited by Rajeev K. Tyagi, Prakriti Sharma and Praveen Sharma

Chapter metrics overview

81 Chapter Downloads

View Full Metrics

Abstract

Pregnancy is a particular physiologic stage during which immune regulation is essential. A successful placentation and subsequent fetal development depend on the delicate balance between moderate pro-inflammatory response and immune tolerance. Findings have pointed out a crucial role for regulatory B cells (Bregs) in establishing an immunomodulatory (IM) environment relevant to pregnancy. In a steady state, Bregs represent 10% of B cells in peripheral blood, a proportion that increases during pregnancy, with the highest rate being observed in post-partum. In the context of pregnancy, Bregs seem to be well positioned to perform the mechanisms that accommodate the growing semi-allogenic fetus and also allow the adequate immune response to pathogen. This chapter discusses the mechanism of action of Bregs during human pregnancy. Also, we will evoke interactions between maternal immune cells and fetal annexes that result in hijacking the naïve B cells to educate and to differentiate them into Bregs.

Keywords

  • pregnancy
  • immunoregulation
  • exosomes
  • cytotrophoblast
  • Bregs

1. Introduction

Preeclampsia (PE) is a placental disorder affecting 2–8% of pregnancies with the highest burden observed in poor countries [1, 2, 3, 4]. PE and severe PE are characterized by exacerbated pro-inflammatory (PI) responses, leading to significant maternal and perinatal morbidity and mortality [5, 6, 7, 8]. Successful placentation and the subsequent fetal development depend on the delicate balance between moderate PI response and immune tolerance.

Recent data has shown that the B cell profile is changed during pregnancy to accommodate the growing fetus [9, 10]. Findings have pointed out a crucial role for regulatory B cells (Bregs) in establishing an immunomodulatory (IM) environment relevant for pregnancy [11]. In a steady state, Bregs represent 10% of B cells in peripheral blood, a proportion which increases during pregnancy, with the highest rate being observed in post-partum [12, 13]. Studies have demonstrated an IM function for IL-10-producing Bregs against ongoing inflammatory events to both limits the infection [14] and promote a successful outcome of pregnancy [15, 16]. It was also shown that early transfer of Bregs from normal pregnant to abortion-prone mice prevented fetal rejection and restored pregnancy tolerance [17].

Indeed, the maternal immune system needs to recognize and accommodate a developing semi-allogeneic fetus. Regulatory T cells are shown to contribute to normal pregnancy, and considering the immune regulatory involvement of Bregs cells in the fields of autoimmunity, transplantation tolerance, and cancer biology, the mechanism underlying Bregs activities during pregnancy needs to be unraveled [18, 19, 20]. The immune regulatory function of Bregs consist of inhibition of the differentiation of effector T cells and dendritic cells (DCs), and activation of Tregs [21, 22].

Today, the consensus is not fully established on the characterization of Bregs with respect to cell surface markers. Recent studies have shown that the regulative roles of Bregs are due to the production of the antiinflammatory cytokine interleukin-10 (IL-10) [23, 24, 25]. However, recent data indicated that some B cell subsets perform regulatory functions without IL-10 involvement suggesting that other Bregs use multimechanistic to regulate immune responses.

In mice, multiple B cell subsets are identified to play regulatory function and include the marginal-zone B cells, the transitional 2 marginal-zone precursor cells, follicular B cells, CD5+CD178+ killer B cells, plasma cells, plasmablasts, CD5+CD1dhiIL-10+ B cells, CD5+B-1a cells, GIFT-15 B cells, TIM-1+ B cells, and PD-L1hi B cells [26, 27]. The IL-10-producing Bregs, also called B10 cells have the CD1dhiCD5+ phenotype [28].

In humans, immature B cells, IL-10+ B cells (B10), GrB+ B cells, Br1 cells, and plasmablasts are identified to have immunosuppressive functions [26]. Previous data in humans have described Bregs as CD19+CD24hiCD27+ [29], which are analogous to the mouse B10 cells [26] and CD19+CD24hiCD38hi cells [26] with the ability to produce IL-10 and to express CD80 and CD86 costimulation molecules [30].

This disparity of these regulatory cells suggests that Bregs are not derived from one specific lineage; rather they may become Bregs following exposure to environmental stimuli such as placenta derived-exosomes.

As PE is PI disease and syncytiotrophoblast (STB)-derived exosomes (SDE) contribute to materno-foetal immuno-tolerance, it will be relevant to understand how STB cells and SDE contribute to PE by altering Bregs differentiation and function during human pregnancy. We will discuss whether a disrupted balance of Bregs could increase susceptibility to PE.

Advertisement

2. Cytotrophoblast cells and placenta-derived exosomes in successful placentation and fetal development

The successful pregnancy requires the suitable development of embryo and adequate placentation [31, 32, 33]. The appropriate placentation is based on the proper capacity of villous cytotrophoblasts to fuse and form the syncytiotrophoblast which contributes to development of placenta. Therefore, abnormal cytotrophoblast differentiation results in placental-related pregnancy diseases [34, 35, 36].

Many cytotrophoblast cell subtypes (cytotrophoblasts (CTBs), extravillous cytotrophoblasts (EVTs) and syncytiotrophoblast (STB)) with different structures and functions are involved in placentation [37, 38]. After implantation of the zygote, trophoblast cells develop from the outer cells which form the wall of the blastocyst, and differentiate into either villous or extravillous trophoblast cells [38]. The STBs are the outer lining of the placenta; fulfill a vast range of role including gas and nutrient exchange between mother and fetus. The trophoblast cell subtypes in addition to secreting hormones and proteins, physically protect the fetus from pathogens [39, 40]. EVTs are invasive trophoblast cells that are important for implantation of the placenta and the development of the fetus [41, 42, 43]. Many pregnancy diseases such as PE and intrauterine growth retardation (IUGR) are the consequences of defective placentation [34, 35, 36]. Therefore, the normal development of the placenta is based on complex mechanisms of proliferation and differentiation of trophoblast cells [44, 45]. Many factors (e.g., interferon-induced transmembrane protein 1 (IFITM) and Storkhead box 1 (STOX1) SNPs, Syncytins, and factors released by placenta soluble fms-like tyrosine kinase-1 (sFlt-1), placenta growth factor (PlGF), transforming growth factor-β (TGF-β)) govern the regulation of cytotrophoblast cell differentiation showing their potential use as biomarker [46, 47, 48].

Placental syncytialization is maintained throughout pregnancy by the fusion of adjacent CTBs [49, 50] and is important for successful pregnancy [49, 51, 52, 53, 54]. Syncytins are important players during syncytialization and Vargas et al. and Lokossou et al. indicated that insufficient Syncytin-2 (Syn-2) expression could be the potential cause of PE, shedding light on the correlation between Syn-2 level and cytotrophoblasts fusion [46, 50, 55, 56].

In recent years, the role of exosomes in the development of the placenta has become more and more precise [46, 56, 57]. These microvesicles with diameters of 20–130 nm are extracellular secreted vesicles and are involved in cell-to-cell communication [58]. They, therefore, affect cytotrophoblast differentiation and immune regulation, especially during pregnancy [58, 59]. Exosomes are secreted by most cells and embedded in various substances including proteins [46, 56, 60, 61], mRNA and miRNA [62], and DNA [63]. They can be transported to distant organs and are thought to modify various cells and organ functions [56, 64, 65]. SDE which embedded placenta-specific molecules, including Syn-2, were involved in CTB fusion [55], embryo implantation via the promotion of T regulatory cells, suppression of Nuclear Factor-kB signaling pathway [66] and thereby in immune reaction and inflammatory response [56]. Secreted exosomes from the placenta into the systemic circulation lead in multisystemic organ damage, in patients with PE [67]. Reduction of Syn-2 levels in exosomes is suggested to be an early biomarker of PE [46, 60]. Indeed, the identification of women at high risk of PE before its onset is especially a challenge. Exosomes miRNA pattern also appears to be used for early PE diagnosis [68]. SDE in preeclamptic placentas are thought to embed high concentrations of PE-specific contents, resulting in unfavorable microenvironments for the invasion of EVTs and the remodeling of spiral arteries for adequate placentation [46, 57, 63, 66, 67, 68, 69, 70, 71, 72].

Nowadays, evidence suggests that disruption of placentation characterizes the pathogenesis of PE [55, 73]. Indeed, STB-derived exosomes are found in maternal circulation [72], and affected endothelial function due to their abundant sFlt-1 and soluble endoglin (sEng) content [69]. These vesicles are also endowed with immune regulation capacities during pregnancy due to Syn-2 embedded in exosomes [56].

Placental exosomes are therefore able to deliver many molecules including proteins around CTB, inducing a particular environment that affects placenta and fetal growth.

The immunosuppressive protein derived from human endogenous retrovirus sequences, Sync-2. plays a leading role in placenta formation [49, 50]. For several years our knowledge has grown on PE and placental exosomes. We have demonstrated the role of Sync-2 in placentation and in T cell immunosuppression [56, 60, 74] during normal pregnancy and PE, suggesting that Sync-2 could be used as an early biomarker of PE. Our recent data from Benin show a gradual diminution, between 7 and 10 weeks of pregnancy (WP), in the incorporation of Sync-2 in serum-derived exosomes from women who had developed a PE later during their pregnancy in comparison to samples from women with normal pregnancy [46]. Sync-2 through its immunosuppressive domain might contribute greatly to creating an immunosuppressive environment. This environment is reinforced and maintained by other factors such as Bregs. This immunosuppressive environment is essential at the beginning of pregnancy for the allograft tolerance constituted by the fetus [75]. As we demonstrated that Sync-2 generates an immunosuppression (IS) environment, Bregs should be important to maintain the IS environment and to prevent allograft rejection. Indeed, PE is a placental and inflammatory disease and syncytiotrophoblast-derived exosomes contribute to materno-fetal immuno-tolerance [56]. Such defective placentation is thought to be caused by an abnormal CTB fusion due to defective production of Sync-2 [46, 49, 55] but also to abnormal maternal immune regulation, involving Sync-2 [56]. Many immune cells, including T cell, macrophage, natural killer, regulatory B and T cells, are also affected during PE [76]. Therefore, it will be of great importance to understand how cytotrophoblast and/or syncytiotrophoblast cells and placenta-derived exosomes contribute to PE by altering Bregs differentiation and function during human pregnancy. By demonstrating that Bregs frequency and function increase susceptibility to PE, would lead to the immediate management of pregnant women predisposed to the development of severe PE and reduce the number of resulting morbidity and deaths.

Advertisement

3. Preeclampsia: state of knowledge

Preeclampsia (PE) is the most common placental disorder affecting pregnancy [77]. PE is associated with vascular dysfunction and deregulated inflammation, oxidative stress, and endothelial dysfunctions [78, 79, 80]. This chronic inflammation begins early in pregnancy as a result of stimulation of maternal immune response by trophoblasts and trophoblasts derived-products. It is associated with leukocyte activation, vascular activation and dysfunction and high serum levels of cytokines such as Tumor Necrosis Factor-α (TNF-α) [81, 82, 83]. Changes in the levels of immune factors (e.g., cytokines, chemokines) are followed by changes in blood coagulation factors, and apoptotic markers [78, 84, 85, 86, 87]. Leukocyte activation is driven by TNF-α, and monocyte-derived cytotoxic protein, that also induces vascular endothelial adhesion molecules. Indeed, increased TNF-α levels in early pregnancy can increase the expression of intercellular adhesion molecule-1 (ICAM-1) on vascular endothelial cells (ECs) and trophoblasts, thereby activating them. Consequently, coagulation cascade, vascular tone, and permeability are disturbed. Moreover, chronic inflammation also activates lymphocyte function-associated antigen-1 (LFA-1) on leukocytes, resulting in the above consequences [84, 85, 88, 89]. This chronic inflammation is also associated with oxidative stress [90] which increases the adhesion of leukocytes to the vascular endothelium and the release of cytokines and anti-angiogenic molecules. Adhesion molecules (e.g., soluble E-selectin and soluble ICAM-1) and reactive oxygen species level are increased in blood collected early in pregnancy from women who develop later PE [83, 91]. During PE, abnormal levels of anti-vascular growth factors (e.g., sFlt-1 and sEng)) lead to maternal vascular inflammatory syndrome characteristics [58]. These antiangiogenic factors induce the decrease of angiogenic placental growth factor (PlGF), poorly affecting angiogenesis during placentation [92]. Defective placenta secreted PlGF into the maternal circulation as a result of impaired endovascular invasion by trophoblast cells and is underlying by cellular oxidative or endoplasmic reticulum stress [93]. Therefore, the level of these factors in the maternal peripheral blood might enable an early diagnosis of PE. Nevertheless, these methods are questioned and did not allow an early prediction (i.e., during the first trimester) of the occurrence of PE [94, 95].

Commonly, PE results in multi-organ failure (e.g., renal insufficiency, liver dysfunction, neurological or hematological complications, uteroplacental dysfunction) in the mother and poor perinatal outcome. Thereby, PE results in significant maternal and perinatal morbidity and mortality [77]. PE affects 2 to 8% of pregnancies and low and middle-income countries (LMIC) are mostly affected [1, 2, 3]. PE is defined as new onset hypertension arising after 20 weeks’ gestation, but can also occur at a later stage, i.e., 4–12 weeks postpartum [96, 97].

PE is a consequence of failure of paternal antigen-specific tolerance. Moreover, first pregnancy, first pregnancy after partner change, and long interval between pregnancies increase PE risk [98, 99]. The reduced opportunities for exposure to seminal plasma, and pregnancy by sperm or oocyte donation or pregnancies with donated greatly increase PE risk [100, 101, 102, 103].

Advertisement

4. Immune regulation and immune tolerance during pregnancy

Contrary to what would be expected, the maternal immune system does not reject the semi-allogeneic fetus allowing the maintenance of the pregnancy [104], but still reacting with infectious agents. This is the result of interplay between maternal and fetal cells creating a tolerogenic microenvironment at the feto-maternal interface [105]. During PE, immunotolerance to fetal antigens, (e.g. trophoblast) is impaired, resulting in disrupted remodeling of the spiral artery and thereby poor placentation. Regulatory T (Tregs) cells play a crucial role in this tolerogenic microenvironment [106, 107] and the maintenance of pregnancy depends on the balance between Tregs and cytotoxic T cells. The ability of fetal cells to escape destruction by maternal immune cells is based on the expression of human leukocyte antigen (HLA)-C molecules by EVTs alone. Therefore, maternal T-cell reactivity to fetal cells is reduced [18]. The T cells immunosuppression during pregnancy is also mediated by HLA-G, E, F and programmed cell death ligand 1 (PD-L1), indoleamine 2,3-dioxygenase (IDO) expression by EVTs [18, 19, 20, 108, 109, 110]. EVTs also induce T cell suppression by expressing cytokines including IL-10, TGF-β, and IL-35 [12, 13]. At the beginning of pregnancy, CD56brightCD16 decidual natural killer (NK) cells (dNK cells) represent more than 60% of decidual immune cell and express high level of immunosuppressive receptors [111, 112]. Immunotolerance is maintained at the feto-maternal interface by interaction between HLA-G expressed on EVTs and dNK cells and dNK derived-cytokines [57, 113, 114]. To maintain immunotolerance at the feto-maternal interface, between dendritic cells (DCs) and Tregs [115], the cross-presentation of paternal antigens to maternal cytotoxicity T CTLs and CD4+ T cells [115, 116] is altered by fetal antigen-specific Tregs at the feto-maternal interface [117, 118, 119, 120, 121]. Seminal plasma components such as TGF-β, prostaglandins, MHCs, and minor antigens also functionally affect maternal antigen-presenting cells (APC). These functional changes were maintained by EVTs [103, 122]. In mice, these functional changes favor fetal-antigen-specific Tregs cell expansion in the uterus and uterine drainage lymph nodes [123, 124, 125]. Moreover, in mice, PD-L2-expressing dendritic cells (DCs) increase during implantation in allogeneic pregnancy in mice [126]. These cells limit inflammation whereas, the decrease of M2 macrophages, which inhibit inflammation and promote tissue repair, results in implantation failure in mice [127]. Furthermore, in vitro, the close interaction between decidual macrophages and EVTs results in an increased number of Tregs after co-culture with peripheral blood-derived CD4+ T cells [128129]. These results show clearly that EVTs oriented the differentiation of CD4+ conventional T cells into antigen-specific peripheral Tregs [121].

Miscarriages, PE, and implantation failure are some characteristics of pregnancy complications. The dysfunction of Tregs is clearly involved [121, 130]. Indeed, in recurrent pregnancy loss and during PE, low level of Tregs in the peripheral blood and uterus has been reported [121]. A recent study has shown that during PE, clonally expanded effector Tregs were significantly decreased in the decidua compared with normal pregnancy, suggesting an insufficient Tregs antigen-specific tolerance [131].

In addition to this induced-immunosuppression, decidual CTLs were also suppressed by EVTs and other immune cells; to allow a good course of pregnancy, without suppression of CTLs functions against virus [117]. During late gestation, le level of PD-L1 on clonally expanded CTLs increases significantly compared to the beginning of pregnancy, showing that strong suppressive signals are necessary to inhibit the allo-reaction by CTLs in the late gestational period [119]. Moreover, during late onset of PE, the level of PD-L1 on clonal CTLs decreased compared with that in normal pregnancy [119], suggesting insufficient suppression of antigen-specific CTLs in PE.

Overall, during normal pregnancy although Tregs and CTLs recognize fetal antigens at the feto-maternal interface, antigen-specific Tregs induce tolerance, while the cytotoxic function of CTLs is suppressed. Therefore, the imbalance of suppressive role of Tregs and activation of CTLs is likely associated with PE.

In PE, type-1 T helper (Th1) cells numbers are also increased [73] and secrete pro-inflammatory cytokines, such as TNF-α, interferon-γ (IFN-γ), and interleukin (IL)-6. Increased Th17 cells secreting the pro-inflammatory cytokine IL-17 are also found during PE [83, 132].

Advertisement

5. Regulatory B cells establish immunomodulatory environment for pregnancy

With regard to their multi-faceted roles, B cells may participate in successful pregnancy [11]. A subtype of B cells named Bregs exhibits immunosuppressive function and therefore is considered an important players in immunological tolerance during pregnancy. During pregnancy, the change of the maternal immune response is governed by a range of cytokines that shape the type and abundance of leukocyte subsets in the decidua and placenta. In addition, these changes also include the reduction of antigen-presenting capacities of monocytes, macrophages, and DCs; inhibition of NK cells, T cells, and B cells; proliferation of dNK cells; maintenance of tolerogenic DCs; and the induction of Tregs [133].

Regulatory roles of Bregs were attributed exclusively to the production of the anti-inflammatory cytokine interleukin-10 [23, 29, 134], even recent data have identified other B cell subsets with regulatory functions without IL-10 production. Indeed, Bregs are cells that dampen ongoing inflammatory events in murine models [15] and counteract excessive pro-inflammatory responses during infection [14]. IL-10 is crucial for optimal pregnancy outcomes, Therefore IL-10 deficiency is related to fetal resorption, growth restriction, and even death of mother and child [135, 136]. This antiinflammatory cytokine is found in high levels in the decidual and placenta during pregnancy and is involved in damping the pro-inflammatory cytokine response. Interesting fact, at the beginning of pregnancy, the inflammation induced by the recognition of paternal antigens is upset by the production of IL-10 [137].

Bregs proportion increases during pregnancy and in postpartum [138] and first-trimester peripheral blood-derived Bregs are shown to inhibit TNF-α secretion by activated T effector cells [139]. In the context of pregnancy, Bregs seem to be well positioned to perform the mechanisms that accommodate the growing semi-allogeneic fetus and also allow the adequate immune response to the pathogen [10]. However, the mechanism of action of Bregs during pregnancy remains curtailed even if their importance in pregnancy has been shown in mouse models [138].

In order to allow a successful placentation and suitable fetal development, the maternal immune system undergoes several changes while allowing the mother to defend herself against infections [10]. A German group has developed a PE mouse model by transferring activated Th1-like splenocytes into normal pregnant mice and has demonstrated that pregnancy-associated immuno-regulation involved a shift from inflammatory toward anti-inflammatory immune responses mainly controlled by T and B cells [140, 141]. They have reported that Bregs were active players in the maintenance of pregnancy by modulating T cell functions [142]. Indeed, they have shown that Bregs transfer from normal pregnant to abortion-prone mice early in pregnancy prevents fetal rejection and restores pregnancy tolerance in mice [17].

The action of Bregs during pregnancy is interconnected with that of Tregs and DCs, providing an appropriate environment for fetal growth.

As described above, both Tregs and DCs play critical roles in determining pregnancy outcomes. In normal pregnancy, fetal-tolerant involves decidual DCs that remain immature and knew as tolerogenic DCs [143]. Tregs are also crucial players in maintaining maternal-fetal immune tolerance. At the onset of embryo implantation, expansion of the Tregs population improves the outcome of pregnancy whereas deficiency or low numbers of Tregs in the uterus during pregnancy leads to pregnancy lost (i.e., abortion or miscarriage) [22, 144].

As anti-inflammatory signal such as IL-10 or TGF-β is crucial to maintain the DC immature phenotype that prevents the activation of T cells [145], IL-10 and or TGF-β must then be initially: 1- present in the uterus and placenta to either drive the induction of immune tolerant DCs followed by the induction of Tregs, or 2- directly drive the induction of Tregs.

Overall Breg’s role in pregnancy seems to be upstream to that of Tregs [17] and therefore, Bregs seem to be the first sources of IL-10 and TGF-β [10, 17].

Advertisement

6. Impact of cytotrophoblast cells and placenta-derived exosomes on regulatory B cells differentiation and function

The pathophysiology of PE is poorly understood despite the evidences supporting a role of immune system in the development of PE [56]. B cells represent a dominant component in the pathogenesis of PE and studies focused on the number and functions of Bregs during PE are of great interest in understanding the pathophysiology of PE [139]. Harnessing Bregs functions may lead to the capacity of using Bregs as an immunotherapeutic agent for averting and treating pregnancy pathologies such as PE. Perinatal cells including cells from term placenta and fetal annexes (amniotic membrane, chorionic membrane, umbilical cord, etc) are able to inhibit B cell proliferation, impair B cell differentiation and promote Bregs formation, frequently due to bioactive factors secreted by perinatal cells [11, 146]. These cells are considered as a promising tool for therapeutic approaches in PE [146]. Interactions between maternal immune cells and fetal annexes may result in hijacking naïve B cells and educating them to become Bregs. However, how cytotrophoblast (CT) and/or syncytiotrophoblast (ST) cells regulate Bregs differentiation and function during pregnancy is still unknown. Maybe in case of PE, CT and ST and their derived-vesicles (e.g. exosomes) will prevent adequate Bregs development and function, resulting in reduced and dysfunctional Bregs. This default of Bregs might result in an inflammatory environment, which will increase the susceptibility to PE.

Recent in vitro and in vivo studies have shown that perinatal cells and perinatal cells derived-vesicles interfere with the activation and differentiation of innate and adaptive immune system cells [11]. Poor knowledge is available about the impact of perinatal cells on B lymphocytes, even if some of the complex cross-talks between perinatal cells and B cells have been described. These studies demonstrated that perinatal cells have a strong antiproliferative capacity on B cells, but were not based on cell–cell contact. The demonstration is based on bioactive factors secreted by perinatal cells. For instance, co-cultured human mesenchymal stromal cells (MSC) isolated from umbilical cord (hUC-MSC) in a contact independent with mouse splenic B cells result in abrogation of the proliferation of activated B cells [147]. Likewise, human umbilical cord matrix cells co-cultured with a B cell cancer line (i.e. Burkitt’s lymphoma cell line) [148], or with auto-reactive B cells from PBMC of immune thrombocytopenic patients results in inhibition of these B cells proliferation [149]. These observations were confirmed by using other perinatal cells (e.g., mesenchymal stromal cells (MSC)) purified from the amniotic membrane (hAMSC). This MSC supernatant is able to suppress CD19+ B cell proliferation in PBMC or purified B cells from PBMC, confirming that cell-to-cell contact was not required and suggesting the role of soluble molecules and vesicles such as exosomes [11]. Similarly, human amniotic fluid stromal cells and their conditioned medium (CM) strongly suppress B cell activation and proliferation, and significantly inhibited the expression of CD80/CD86 costimulatory molecules on activated B lymphocytes [150].

However, some data contradict these observations and showed that human amniotic fluid stromal cells are able to suppress the apoptosis of B lymphocytes, favoring an increase in activated B cell survival. The mechanism underlying this inhibition is based on the decrease of the expression of the negative co-inhibitory molecules B7 homolog 4 (B7H4) and programmed death-ligand 1 (PD-L1) on activated B lymphocytes [150]. Moreover, an increase in B cell proliferation and a reduction in spontaneous apoptosis in the presence of human amniotic epithelial cells (hAEC) were also described [143]. Umbilical cord derived-MSC were not able to affect [144] or in other studies able to highly induce the in vitro growth of PBMC derived-B cells [145].

It is also demonstrated that human amniotic fluid stromal cells induce down-regulation of the proportion of B1 cells [150], resulting in the reduction of the B cell subset mainly involved in the production of autoantibodies in PE [151, 152, 153]. Many studies have shown that perinatal cell and their CM are able to block antibody-secreting cells CD19+CD27+CD38+ and the differentiation of B cells into CD138+ plasma cells, resulting in the reduction of secreted immunoglobulin [11, 147, 150]. However, co-culture of purified B cells with human amniotic fluid stromal cells results in reduction of the proportion of CD19+CD20+CD27+ memory B cells [150], whereas PBMC cultured in the presence of CM-hAMSC increases CD19+CD27+CD38 memory B cells [11]. These different results may be explained by the presence of other immune cells among PBMC instead purified B cells. Moreover, different conditions of stimulation were used to activate B cells, and the lack of consensus in the markers used to characterize the B cell population could also support the distinct results observed by different groups.

Perinatal cells not only modulate B cell function by favoring their differentiation toward plasma cells, but they also promote the formation of Bregs. Indeed, it was reported that hAEC induced the expansion of CD19+CD24hiCD38hi Bregs [143]. However, recent data suggested that IL-10+ Bregs were inhibited by human amniotic fluid stromal cells [150]. These observations clearly showed that more knowledge is needed to understand the impact of perinatal cells and other related vesicles on Bregs differentiation and functions. Thereby, it’s important to identify the signaling pathways involved in underlying how perinatal cells and derivatives affect B cell proliferation and differentiation. Two signaling pathways were identified to be suppressed through CpG oligodeoxynucleotides (CpG ODN) by hAMSC: 1-the Toll-like receptor 9 (TLR9)-myeloid differentiation primary response 88 (MyD88)-interleukin-1 receptor-associated kinase (IRAK)1/4 and 2- the TLR9-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) pathways [11]. This suppression results in a reduction of uptake of the CpG ODN by CD205, TLR9, and CD14. Consequently, IRAK-4, mitogen-activated protein kinases (MAPK) (c-Jun N-terminal Kinase (JNK), p38 MAPK, extracellular signal-regulated kinase (ERK)) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) pathways were inhibited. This induces an important reduction in the expression of phosphorylated AKT [11, 147]. The exact mechanism by which perinatal cells and derivatives induce Bregs differentiation is still unknown, and needs to be investigated [154]. Few data demonstrate that perinatal cells produce soluble factors including prostanoids (i.e., prostaglandin E2 (PGE2)), and maybe exosomes to immune regulate cells [155, 156, 157]. Therefore, we can speculate that Bregs differentiation is also induced by bioactive vesicles.

Based on in vitro results showing that perinatal cells have immunomodulatory properties, they were successfully tested in several inflammatory and immune-mediated diseases, including lung [158, 159] and liver [160] fibrosis, inflammatory bowel disease, collagen-induced arthritis, experimental autoimmune encephalomyelitis [161], multiple sclerosis, wound healing [157, 162], traumatic brain injury [163], cerebral ischemia [164], Huntington’s disease [165], and diabetes [166].

The therapeutic using hAMSC in pathological conditions driven by B cells has demonstrated a reduced idiopathic pulmonary fibrosis progression [158]. This treatment allows low levels of B cells in alveolar spaces and reduced the amount of CD138+ antibody-secreting cells in lung tissues, suggesting a decrease in B cell recruitment and an impairment of the maturation of B cells. Therapy using hAEC has also shown remarkable results in animal models of Hashimoto’s thyroiditis and systemic lupus erythematosus (SLE) [167].

hAEC induced significant up-regulation of Bregs in experimental autoimmune thyroiditis mice. In this experiment, authors have shown that B10 cells are the major target of hAEC. In SLE mice, hAEC has shown the reduction in autoantibody production but without effect on B10 cells, suggesting that the mechanism of hAEC immunomodulation depends on the disease [167].

In the context of chronic graft-versus-host disease (cGVHD) prophylaxis repeated infusion of hUC-MSC seems to minimize the severity and the symptoms of cGVHD by increasing CD27+ memory B cells [168].

The immune modulation properties of perinatal cells depend on the origin of these cells. Indeed, fetal-derived cells induce strong inhibition of T-cell proliferation, cytotoxicity, and switch to M2 macrophages, while maternal-derived cells were more strongly able to induce Tregs [169]. Figure 1 describes the probable implication of exosomes in Bregs differentiation and function (Figure 1).

Figure 1.

Differentiation and functional properties of Bregs. Through the production of exosomes by fetal annexes including cytotrophoblast cells, naive B cells can differentiate into Bregs. By producing IL-10, TGF-b, and IL-35, Bregs can suppress tumor necrosis factor-a (TNF-a)-producing monocytes, IL-12-producing dendritic cells, Th17 cells, Th1 cells, and cytotoxic CD8+ T cells. Bregs can also induce the differentiation of immunosuppressive Tregs, T regulatory 1 (Tr1), and dNK cells. This figure was created using Biorender.com.

As PE is pro-inflammatory disease and syncytiotrophoblast-derived exosomes (SDE) contribute to materno-fetal immuno-tolerance, it will be useful to understand how STB cells and SDE contribute to PE by altering Bregs differentiation and function during human pregnancy. These mechanisms may be close to those that inhibit immune flares or chronic inflammation in autoimmune diseases and transplantation.

Advertisement

7. Conclusion

Gestation is a remarkable biological process in which the mother carries a fetus harboring half of a foreign genome belonging to the father. To allow the growth of the fetus, the maternal immune system needs to accommodate the semi-allogeneic fetus by dampening its immune responses. This results in a state of immunological tolerance throughout gravidity while maintaining the capacity to respond to pathogens properly. This paradoxical situation requires a perfect regulation of the balance between immune tolerance and immune activation.

To enable more accurate prediction and prevention of PE, its pathogenesis needs to be more understood. Increasing evidence suggests a consequence of the altered immune system in the development of PE. Today, it is clear that perinatal cells have capacity to regulate B cell response at different levels: by inhibiting B cell multiplication, impairing B cell differentiation, and inducing B regulatory cell formation. Future research should focus on understanding how cytotrophoblast cells and placenta-derived exosomes act on B cells.

Overall, it is clear that cytotrophoblast cells and placenta-derived exosomes harbor the capacity of being a novel therapeutic approach for PE. However, the opposite results and the mainly small number of studies exploring the effect of cytotrophoblast cells and placenta-derived exosomes on the Bregs subset cannot allow deciding on a position. Further in vitro and in vivo studies are necessary to better decide the immunomodulatory potential of perinatal cells, leading to an important strategy for the treatment of PE.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Steegers EAP, von Dadelszen P, Duvekot JJ, Pijnenborg R. Pre-eclampsia. Lancet. 2010;376(9741):631-644
  2. 2. Maher GM, O’Keeffe GW, Kenny LC, Kearney PM, Dinan TG, Khashan AS. Hypertensive disorders of pregnancy and risk of neurodevelopmental disorders in the offspring: A systematic review and meta-analysis protocol. BMJ Open. Oct 2017;7(10):e018313
  3. 3. McLaren ZM, Sharp A, Hessburg JP, Sarvestani AS, Parker E, Akazili J, et al. Cost effectiveness of medical devices to diagnose pre-eclampsia in low-resource settings. Development Engineering. 2017;2:99-106
  4. 4. von Dadelszen P, Magee LA. Pre-eclampsia: An Update. Current Hypertension Reports. 2014 [cité 28 déc 2017];16(8):454-468. DOI: 10.1007/s11906-014-0454-8
  5. 5. Chaiworapongsa T, Chaemsaithong P, Yeo L, Romero R. Pre-eclampsia part 1: Current understanding of its pathophysiology. Nature Reviews Nephrology. 2014;10(8):466-480
  6. 6. Hsu P, Nanan RKH. Innate and adaptive immune interactions at the fetal and maternal Interface in healthy human pregnancy and pre-eclampsia. Frontiers in Immunology. 2014 [cité 26 janv 2018];5:1-12. DOI: 10.3389/fimmu.2014.00125/abstract
  7. 7. Luppi P, Tse H, Lain KY, Markovic N, Piganelli JD, DeLoia JA. Preeclampsia activates circulating immune cells with engagement of the NF-kappaB pathway. American Journal of Reproductive Immunology. 2006;56(2):135-144
  8. 8. Luppi P, DeLoia JA. Monocytes of preeclamptic women spontaneously synthesize pro-inflammatory cytokines. Clinical Immunology. 2006;118(2-3):268-275
  9. 9. Muzzio DO, Soldati R, Ehrhardt J, Utpatel K, Evert M, Zenclussen AC, et al. B cell development undergoes profound modifications and adaptations during pregnancy in Mice1. Biology of Reproduction. 2014 [cité 7 avr 2022];91(5):29-37. DOI: 10.1095/biolreprod.114.122366
  10. 10. Guzman-Genuino RM, Diener KR. Regulatory B cells in pregnancy: Lessons from autoimmunity, graft tolerance, and Cancer. Frontiers in Immunology. 2017 [cité 7 avr 2022];8:1-19. DOI: 10.3389/fimmu.2017.00172/full
  11. 11. Magatti M, Masserdotti A, Cargnoni A, Papait A, Stefani FR, Silini AR, et al. The role of B cells in PE pathophysiology: A potential target for perinatal cell-based therapy? IJMS. 2021;22(7):3405
  12. 12. Dar HY, Rani L, Sapra L, Azam Z, Shokeen N, Bhardwaj A, et al. A chronological journey of Breg subsets: Implications in health and disease. In: Singh S, editor. Systems and Synthetic Immunology. Singapore: Springer Singapore; 2020 [cité 8 sept 2022]. pp. 125-152. DOI: 10.1007/978-981-15-3350-1_5
  13. 13. Busse M, Campe KNJ, Redlich A, Oettel A, Hartig R, Costa SD, et al. Regulatory B cells are decreased and impaired in their function in peripheral maternal blood in pre-term birth. Frontiers in Immunology. 2020;11:386
  14. 14. Boldison J, Da Rosa LC, Davies J, Wen L, Wong FS. Dendritic cells license regulatory B cells to produce IL-10 and mediate suppression of antigen-specific CD8 T cells. Cellular & Molecular Immunology. 2020;17(8):843-855
  15. 15. Busse M, Campe KNJ, Nowak D, Schumacher A, Plenagl S, Langwisch S, et al. IL-10 producing B cells rescue mouse fetuses from inflammation-driven fetal death and are able to modulate T cell immune responses. Scientific Reports. 2019;9(1):9335
  16. 16. Rolle L, Memarzadeh Tehran M, Morell-García A, Raeva Y, Schumacher A, Hartig R, et al. Cutting edge: IL-10-producing regulatory B cells in early human pregnancy. American Journal of Reproductive Immunology. 2013;70(6):448-453
  17. 17. Jensen F, Muzzio D, Soldati R, Fest S, Zenclussen AC. Regulatory B10 cells restore pregnancy tolerance in a mouse Model1. Biology of Reproduction. 2013 [cité 20 mai 2020];89(4):1-7. DOI: 10.1095/biolreprod.113.110791
  18. 18. King A, Burrows TD, Hiby SE, Bowen JM, Joseph S, Verma S, et al. Surface expression of HLA-C antigen by human extravillous trophoblast. Placenta. 2000;21(4):376-387
  19. 19. Ishitani A, Sageshima N, Lee N, Dorofeeva N, Hatake K, Marquardt H, et al. Protein expression and peptide binding suggest unique and interacting functional roles for HLA-E, F, and G in maternal-placental immune recognition. Journal of Immunology. 2003;171(3):1376-1384
  20. 20. Kovats S, Main EK, Librach C, Stubblebine M, Fisher SJ, DeMars R. A class I antigen, HLA-G, expressed in human trophoblasts. Science. 1990;248(4952):220-223
  21. 21. Segerer SE, Staib C, Kaemmerer U, Frambach T, Honig A, Dietl J, et al. Dendritic cells: Elegant arbiters in human reproduction. Current Pharmaceutical Biotechnology. 2012;13(8):1378-1384
  22. 22. Ruocco MG, Chaouat G, Florez L, Bensussan A, Klatzmann D. Regulatory T-cells in pregnancy: Historical perspective, state of the art, and burning questions. Frontiers in Immunology. 2014 [cité 8 sept 2022];5:1-10. DOI: 10.3389/fimmu.2014.00389/abstract
  23. 23. Fillatreau S, Sweenie CH, McGeachy MJ, Gray D, Anderton SM. B cells regulate autoimmunity by provision of IL-10. Nature Immunology. 2002;3(10):944-950
  24. 24. Matsushita T, Yanaba K, Bouaziz JD, Fujimoto M, Tedder TF. Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression. The Journal of Clinical Investigation. 2008;118(10):3420-3430
  25. 25. Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, et al. Identification of a B cell signature associated with renal transplant tolerance in humans. Journal of Clinical Investigation. 2010;120(6):1836-1847
  26. 26. Mauri C, Menon M. The expanding family of regulatory B cells. International Immunology. 2015;27(10):479-486
  27. 27. Rosser EC, Mauri C. Regulatory B cells: Origin, phenotype, and function. Immunity. 2015;42(4):607-612
  28. 28. Yanaba K, Bouaziz JD, Haas KM, Poe JC, Fujimoto M, Tedder TF. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity. 2008;28(5):639-650
  29. 29. Mauri C, Gray D, Mushtaq N, Londei M. Prevention of arthritis by interleukin 10-producing B cells. The Journal of Experimental Medicine. 2003;197(4):489-501
  30. 30. Blair LYN, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, et al. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic lupus erythematosus patients. 2010;32(1):129-140. doi: 10.1016/j.immuni.2009.11.009
  31. 31. Hamilton WJ, Boyd JD. Development of the human placenta in the first three months of gestation. Journal of Anatomy. 1960;94:297-328
  32. 32. Castellucci M, Scheper M, Scheffen I, Celona A, Kaufmann P. The development of the human placental villous tree. Anatomy and Embryology (Berl). 1990;181(2):117-128
  33. 33. Jones CJ, Fox H. Ultrastructure of the normal human placenta. Electron Microscopy Reviews. 1991;4(1):129-178
  34. 34. Fisher SJ. Why is placentation abnormal in preeclampsia? American Journal of Obstetrics and Gynecology. 2015;213(4 Suppl):S115-S122
  35. 35. Khong TY, Liddell HS, Robertson WB. Defective haemochorial placentation as a cause of miscarriage: A preliminary study. British Journal of Obstetrics and Gynaecology. 1987;94(7):649-655
  36. 36. Khong TY, De Wolf F, Robertson WB, Brosens I. Inadequate maternal vascular response to placentation in pregnancies complicated by pre-eclampsia and by small-for-gestational age infants. British Journal of Obstetrics and Gynaecology. 1986;93(10):1049-1059
  37. 37. Gude NM, Roberts CT, Kalionis B, King RG. Growth and function of the normal human placenta. Thrombosis Research. 2004;114(5-6):397-407
  38. 38. Xiao Z, Yan L, Liang X, Wang H. Progress in deciphering trophoblast cell differentiation during human placentation. Current Opinion in Cell Biology. 2020;67:86-91
  39. 39. Soilleux EJ, Coleman N. Transplacental transmission of HIV: A potential role for HIV binding lectins. The International Journal of Biochemistry & Cell Biology. 2003;35(3):283-287
  40. 40. Arechavaleta-Velasco F, Koi H, Strauss JF, Parry S. Viral infection of the trophoblast: Time to take a serious look at its role in abnormal implantation and placentation? Journal of Reproductive Immunology. 2002;55(1-2):113-121
  41. 41. Burton GJ, Jauniaux E. The cytotrophoblastic shell and complications of pregnancy. Placenta. 2017;60:134-139
  42. 42. Velicky P, Meinhardt G, Plessl K, Vondra S, Weiss T, Haslinger P, et al. Genome amplification and cellular senescence are hallmarks of human placenta development. PLoS Genetics. 2018;14(10):e1007698
  43. 43. Pijnenborg R, Dixon G, Robertson WB, Brosens I. Trophoblastic invasion of human decidua from 8 to 18 weeks of pregnancy. Placenta. 1980;1(1):3-19
  44. 44. Baczyk D, Drewlo S, Proctor L, Dunk C, Lye S, Kingdom J. Glial cell missing-1 transcription factor is required for the differentiation of the human trophoblast. Cell Death and Differentiation. 2009;16(5):719-727
  45. 45. Renaud SJ, Kubota K, Rumi MAK, Soares MJ. The FOS transcription factor family differentially controls trophoblast migration and invasion. The Journal of Biological Chemistry. 2014;289(8):5025-5039
  46. 46. Gatien L. Low serum-derived Syncytin-2 levels in exosome at early pregnancy is a predictor of preeclampsia: A prospective pilot study in Benin, West Africa. Journal of Obstetrics and Gynecological Surgery. 2020;17:1-6
  47. 47. Nguyen TPH, Patrick CJ, Parry LJ, Familari M. Using proteomics to advance the search for potential biomarkers for preeclampsia: A systematic review and meta-analysis. PLoS One. 2019;14(4):e0214671
  48. 48. Khaliq OP, Konoshita T, Moodley J, Naicker T. The role of uric acid in preeclampsia: Is uric acid a causative factor or a sign of preeclampsia? Current Hypertension Reports. 2018;20(9):80
  49. 49. Lokossou AG, Toufaily C, Vargas A, Barbeau B. siRNA transfection and EMSA analyses on freshly isolated human villous Cytotrophoblasts. Journal of Visualized Experiments. 2016;115:1-9
  50. 50. Vargas A, Moreau J, Landry S, LeBellego F, Toufaily C, Rassart É, et al. Syncytin-2 plays an important role in the fusion of human trophoblast cells. Journal of Molecular Biology. 2009;392(2):301-318
  51. 51. Langbein M, Strick R, Strissel PL, Vogt N, Parsch H, Beckmann MW, et al. Impaired cytotrophoblast cell-cell fusion is associated with reduced Syncytin and increased apoptosis in patients with placental dysfunction. Molecular Reproduction and Development. 2008;75(1):175-183
  52. 52. Huppertz B. Placental origins of preeclampsia: Challenging the current hypothesis. Hypertension. 2008;51(4):970-975
  53. 53. Vargas A, Toufaily C, LeBellego F, Rassart É, Lafond J, Barbeau B. Reduced expression of both Syncytin 1 and Syncytin 2 correlates with severity of preeclampsia. Reproductive Sciences. 2011;18(11):1085-1091
  54. 54. Lu X, Wang R, Zhu C, Wang H, Lin HY, Gu Y, et al. Fine-tuned and cell-cycle-restricted expression of Fusogenic protein Syncytin-2 maintains functional placental syncytia. Cell Reports. 2017;21(5):1150-1159
  55. 55. Vargas A, Zhou S, Éthier-Chiasson M, Flipo D, Lafond J, Gilbert C, et al. Syncytin proteins incorporated in placenta exosomes are important for cell uptake and show variation in abundance in serum exosomes from patients with preeclampsia. The FASEB Journal. 2014;28(8):3703-3719
  56. 56. Lokossou AG, Toudic C, Nguyen PT, Elisseeff X, Vargas A, Rassart É,et al. Endogenous retrovirus-encoded Syncytin-2 contributes to exosome-mediated immunosuppression of T cells. Biology of Reproduction. 2020;102(1):185-198
  57. 57. Matsubara K, Matsubara Y, Uchikura Y, Sugiyama T. Pathophysiology of preeclampsia: The role of exosomes. International Journal of Molecular Sciences. 2021;22(5):2572
  58. 58. Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, et al. Circulating angiogenic factors and the risk of preeclampsia. The New England Journal of Medicine. 2004;350(7):672-683
  59. 59. Bobrie A, Colombo M, Raposo G, Théry C. Exosome secretion: Molecular mechanisms and roles in immune responses. Traffic. 2011;12(12):1659-1668
  60. 60. Lokossou AG, Toudic C, Barbeau B. Implication of human endogenous retrovirus envelope proteins in placental functions. Viruses. 2014;6(11):4609-4627
  61. 61. Kar M. Role of biomarkers in early detection of preeclampsia. Journal of Clinical and Diagnostic Research. 2014;8(4):BE01-BE04
  62. 62. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007;9(6):654-659
  63. 63. Konečná B, Tóthová Ľ, Repiská G. Exosomes-associated DNA-new marker in pregnancy complications? International Journal of Molecular Sciences. 2019;20(12):E2890
  64. 64. Bowers EC, Hassanin AAI, Ramos KS. In vitro models of exosome biology and toxicology: New frontiers in biomedical research. Toxicology in Vitro. 2020;64:104462
  65. 65. Thomou T, Mori MA, Dreyfuss JM, Konishi M, Sakaguchi M, Wolfrum C, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. 2017;542(7642):450-455
  66. 66. Czernek L, Düchler M. Exosomes as messengers between mother and fetus in pregnancy. International Journal of Molecular Sciences. 2020;21(12):E4264
  67. 67. Pillay P, Moodley K, Moodley J, Mackraj I. Placenta-derived exosomes: Potential biomarkers of preeclampsia. International Journal of Nanomedicine. 2017;12:8009-8023
  68. 68. Devor E, Santillan D, Scroggins S, Warrier A, Santillan M. Trimester-specific plasma exosome microRNA expression profiles in preeclampsia. The Journal of Maternal-Fetal & Neonatal Medicine. 2020;33(18):3116-3124
  69. 69. Gill M, Motta-Mejia C, Kandzija N, Cooke W, Zhang W, Cerdeira AS, et al. Placental Syncytiotrophoblast-derived extracellular vesicles carry active NEP (Neprilysin) and are increased in preeclampsia. Hypertension. 2019;73(5):1112-1119
  70. 70. Kshirsagar SK, Alam SM, Jasti S, Hodes H, Nauser T, Gilliam M, et al. Immunomodulatory molecules are released from the first trimester and term placenta via exosomes. Placenta. 2012;33(12):982-990
  71. 71. Mitchell MD, Peiris HN, Kobayashi M, Koh YQ , Duncombe G, Illanes SE, et al. Placental exosomes in normal and complicated pregnancy. American Journal of Obstetrics and Gynecology. 2015;213(4):S173-S181
  72. 72. Pillay P, Maharaj N, Moodley J, Mackraj I. Placental exosomes and pre-eclampsia: Maternal circulating levels in normal pregnancies and, early and late onset pre-eclamptic pregnancies. Placenta. 2016;46:18-25
  73. 73. Saito S, Sakai M. Th1/Th2 balance in preeclampsia. Journal of Reproductive Immunology. 2003;59(2):161-173
  74. 74. Toufaily C, Lokossou AG, Vargas A, Rassart É, Barbeau B. A CRE/AP-1-like motif is essential for induced syncytin-2 expression and fusion in human trophoblast-like model. PLoS One. 2015;10(3):e0121468
  75. 75. Ghaebi M, Nouri M, Ghasemzadeh A, Farzadi L, Jadidi-Niaragh F, Ahmadi M, et al. Immune regulatory network in successful pregnancy and reproductive failures. Biomedicine & Pharmacotherapy. 2017;88:61-73
  76. 76. Miller D, Motomura K, Galaz J, Gershater M, Lee ED, Romero R, et al. Cellular immune responses in the pathophysiology of preeclampsia. Journal of Leukocyte Biology. 2022;111(1):237-260
  77. 77. Chappell LC, Enye S, Seed P, Briley AL, Poston L, Shennan AH. Adverse perinatal outcomes and risk factors for preeclampsia in women with chronic hypertension: A prospective study. Hypertension. 2008;51(4):1002-1009
  78. 78. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA, McLaughlin MK. Preeclampsia: An endothelial cell disorder. American Journal of Obstetrics and Gynecology. 1989;161(5):1200-1204
  79. 79. Roberts JM, Hubel CA. The two stage model of preeclampsia: Variations on the theme. Placenta. 2009;30(Suppl. A):S32-S37
  80. 80. Soleymanlou N, Jurisica I, Nevo O, Ietta F, Zhang X, Zamudio S, et al. Molecular evidence of placental hypoxia in preeclampsia. The Journal of Clinical Endocrinology and Metabolism. 2005;90(7):4299-4308
  81. 81. Abe E, Matsubara K, Ochi H, Ito M, Oka K, Kameda K. Elevated levels of adhesion molecules derived from leukocytes and endothelial cells in patients with pregnancy-induced hypertension. Hypertension in Pregnancy. 2003;22(1):31-43
  82. 82. Abe E, Matsubara K, Oka K, Kusanagi Y, Ito M. Cytokine regulation of intercellular adhesion molecule-1 expression on trophoblasts in preeclampsia. Gynecologic and Obstetric Investigation. 2008;66(1):27-33
  83. 83. Matsubara K, Abe E, Ochi H, Kusanagi Y, Ito M. Changes in serum concentrations of tumor necrosis factor alpha and adhesion molecules in normal pregnant women and those with pregnancy-induced hypertension. The Journal of Obstetrics and Gynaecology Research. 2003;29(6):422-426
  84. 84. Ballegeer VC, Spitz B, De Baene LA, Van Assche AF, Hidajat M, Criel AM. Platelet activation and vascular damage in gestational hypertension. American Journal of Obstetrics and Gynecology. 1992;166(2):629-633
  85. 85. Roberts JM, Taylor RN, Goldfien A. Clinical and biochemical evidence of endothelial cell dysfunction in the pregnancy syndrome preeclampsia. American Journal of Hypertension. 1991;4(8):700-708
  86. 86. Tjoa ML, Jani J, Lewi L, Peter I, Wataganara T, Johnson KL, et al. Circulating cell-free fetal messenger RNA levels after fetoscopic interventions of complicated pregnancies. American Journal of Obstetrics and Gynecology. 2006;195(1):230-235
  87. 87. Taglauer ES, Wilkins-Haug L, Bianchi DW. Review: Cell-free fetal DNA in the maternal circulation as an indication of placental health and disease. Placenta. 2014;35(Suppl):S64-S68
  88. 88. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 1988;332(6163):411-415
  89. 89. Palmer RM, Ashton DS, Moncada S. Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature. 1988;333(6174):664-666
  90. 90. Roberts JM, Hubel CA. Is oxidative stress the link in the two-stage model of pre-eclampsia? Lancet. 1999;354(9181):788-789
  91. 91. Matsubara K, Matsubara Y, Hyodo S, Katayama T, Ito M. Role of nitric oxide and reactive oxygen species in the pathogenesis of preeclampsia. The Journal of Obstetrics and Gynaecology Research. 2010;36(2):239-247
  92. 92. Kifle MM, Dahal P, Vatish M, Cerdeira AS, Ohuma EO. The prognostic utility of soluble fms-like tyrosine kinase-1 (sFlt-1) and placental growth factor (PIGF) biomarkers for predicting preeclampsia: A secondary analysis of data from the INSPIRE trial. BMC Pregnancy and Childbirth. 2022;22(1):520
  93. 93. Burton GJ, Jauniaux E. Placental oxidative stress: From miscarriage to preeclampsia. Journal of the Society for Gynecologic Investigation. 2004;11(6):342-352
  94. 94. Schlembach D, Hund M, Schroer A, Wolf C. Economic assessment of the use of the sFlt-1/PlGF ratio test to predict preeclampsia in Germany. BMC Health Services Research. 6 août 2018;18(1):603
  95. 95. Hodel M, Blank PR, Marty P, Lapaire O. sFlt-1/PlGF ratio as a predictive marker in women with suspected preeclampsia: An economic evaluation from a Swiss perspective. Disease Markers. 2019;2019:4096847
  96. 96. Brown MA, Lindheimer MD, de Swiet M, Van Assche A, Moutquin JM. The classification and diagnosis of the hypertensive disorders of pregnancy: Statement from the International Society for the Study of hypertension in pregnancy (ISSHP). Hypertension in Pregnancy. 2001;20(1):IX-XIV
  97. 97. Phipps E, Prasanna D, Brima W, Jim B. Preeclampsia: Updates in pathogenesis, definitions, and guidelines. Clinical Journal of the American Society of Nephrology. 2016;11(6):1102-1113
  98. 98. Robillard PY, Hulsey TC, Alexander GR, Keenan A, de Caunes F, Papiernik E. Paternity patterns and risk of preeclampsia in the last pregnancy in multiparae. Journal of Reproductive Immunology. 1993;24(1):1-12
  99. 99. Skjaerven R, Wilcox AJ, Lie RT. The interval between pregnancies and the risk of preeclampsia. The New England Journal of Medicine. 2002;346(1):33-38
  100. 100. Salha O, Sharma V, Dada T, Nugent D, Rutherford AJ, Tomlinson AJ, et al. The influence of donated gametes on the incidence of hypertensive disorders of pregnancy. Human Reproduction. 1999;14(9):2268-2273
  101. 101. Melchiorre K, Giorgione V, Thilaganathan B. The placenta and preeclampsia: Villain or victim? American Journal of Obstetrics and Gynecology. 2022;226(2S):S954-S962
  102. 102. Klonoff-Cohen HS, Savitz DA, Cefalo RC, McCann MF. An epidemiologic study of contraception and preeclampsia. Journal of the American Medical Association. 1989;262(22):3143-3147
  103. 103. Koelman CA, Coumans AB, Nijman HW, Doxiadis II, Dekker GA, Claas FH. Correlation between oral sex and a low incidence of preeclampsia: A role for soluble HLA in seminal fluid? Journal of Reproductive Immunology. 2000;46(2):155-166
  104. 104. Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. Nature. 1953;172(4379):603-606
  105. 105. Saito S, Nishikawa K, Morii T, Narita N, Enomoto M, Ichijo M. Expression of activation antigens CD69, HLA-DR, interleukin-2 receptor-alpha (IL-2R alpha) and IL-2R beta on T cells of human decidua at an early stage of pregnancy. Immunology. 1992;75(4):710-712
  106. 106. Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nature Immunology. 2004;5(3):266-271
  107. 107. Saito S, Shiozaki A, Nakashima A, Sakai M, Sasaki Y. The role of the immune system in preeclampsia. Molecular Aspects of Medicine. 2007;28(2):192-209
  108. 108. Nagamatsu T, Schust DJ, Sugimoto J, Barrier BF. Human decidual stromal cells suppress cytokine secretion by allogenic CD4+ T cells via PD-1 ligand interactions. Human Reproduction. 2009;24(12):3160-3171
  109. 109. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. The Journal of Experimental Medicine. 2000;192(7):1027-1034
  110. 110. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281(5380):1191-1193
  111. 111. Koopman LA, Kopcow HD, Rybalov B, Boyson JE, Orange JS, Schatz F, et al. Human Decidual natural killer cells are a unique NK cell subset with immunomodulatory potential. Journal of Experimental Medicine. 2003;198(8):1201-1212
  112. 112. Higuma-Myojo S, Sasaki Y, Miyazaki S, Sakai M, Siozaki A, Miwa N, et al. Cytokine profile of natural killer cells in early human pregnancy. American Journal of Reproductive Immunology. 2005;54(1):21-29
  113. 113. Darmochwal-Kolarz D, Kolarz B, Rolinski J, Leszczynska-Gorzelak B, Oleszczuk J. The concentrations of soluble HLA-G protein are elevated during mid-gestation and decreased in pre-eclampsia. Folia Histochemica et Cytobiologica. 2012;50(2):286-291
  114. 114. Xu X, Zhou Y, Wei H. Roles of HLA-G in the maternal-fetal immune microenvironment. Frontiers in Immunology. 2020;11:592010
  115. 115. Saito S, Shima T, Nakashima A, Inada K, Yoshino O. Role of paternal antigen-specific Treg cells in successful implantation. American Journal of Reproductive Immunology. 2016;75(3):310-316
  116. 116. Murata H, Tanaka S, Tsuzuki-Nakao T, Kido T, Kakita-Kobayashi M, Kida N, et al. The transcription factor HAND2 up-regulates transcription of the IL15 gene in human endometrial stromal cells. The Journal of Biological Chemistry. 2020;295(28):9596-9605
  117. 117. van der Zwan A, Bi K, Norwitz ER, Crespo ÂC, Claas FHJ, Strominger JL, et al. Mixed signature of activation and dysfunction allows human decidual CD8+ T cells to provide both tolerance and immunity. Proceedings of the National Academy of Sciences of the United States of America. 2018;115(2):385-390
  118. 118. Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM, et al. Human decidual tissue contains differentiated CD8+ effector-memory T cells with unique properties. Journal of Immunology. 2010;185(7):4470-4477
  119. 119. Morita K, Tsuda S, Kobayashi E, Hamana H, Tsuda K, Shima T, et al. Analysis of TCR repertoire and PD-1 expression in Decidual and peripheral CD8+ T cells reveals distinct immune mechanisms in miscarriage and preeclampsia. Frontiers in Immunology. 2020;11:1082
  120. 120. Kinder JM, Turner LH, Stelzer IA, Miller-Handley H, Burg A, Shao TY, et al. CD8+ T cell functional exhaustion overrides pregnancy-induced fetal antigen Alloimmunization. Cell Reports. 2020;31(12):107784
  121. 121. Tsuda S, Nakashima A, Morita K, Shima T, Yoneda S, Kishi H, et al. The role of decidual regulatory T cells in the induction and maintenance of fetal antigen-specific tolerance: Imbalance between regulatory and cytotoxic T cells in pregnancy complications. Human Immunology. 2021;82(5):346-352
  122. 122. Robertson SA, Prins JR, Sharkey DJ, Moldenhauer LM. Seminal fluid and the generation of regulatory T cells for embryo implantation. American Journal of Reproductive Immunology. 2013;69(4):315-330
  123. 123. Robertson SA, Guerin LR, Bromfield JJ, Branson KM, Ahlström AC, Care AS. Seminal fluid drives expansion of the CD4+CD25+ T regulatory cell pool and induces tolerance to paternal alloantigens in mice. Biology of Reproduction. 2009;80(5):1036-1045
  124. 124. Shima T, Inada K, Nakashima A, Ushijima A, Ito M, Yoshino O, et al. Paternal antigen-specific proliferating regulatory T cells are increased in uterine-draining lymph nodes just before implantation and in pregnant uterus just after implantation by seminal plasma-priming in allogeneic mouse pregnancy. Journal of Reproductive Immunology. 2015;108:72-82
  125. 125. Shima T, Nakashima A, Yasuda I, Ushijima A, Inada K, Tsuda S, et al. Uterine CD11c+ cells induce the development of paternal antigen-specific Tregs via seminal plasma priming. Journal of Reproductive Immunology. 2020;141:103165
  126. 126. Yasuda I, Shima T, Moriya T, Ikebuchi R, Kusumoto Y, Ushijima A, et al. Dynamic changes in the phenotype of dendritic cells in the uterus and uterine draining lymph nodes after coitus. Frontiers in Immunology. 2020;11:557720
  127. 127. Ono Y, Yoshino O, Hiraoka T, Sato E, Fukui Y, Ushijima A, et al. CD206+ M2-like macrophages are essential for successful implantation. Frontiers in Immunology. 2020;11:557184
  128. 128. Salvany-Celades M, van der Zwan A, Benner M, Setrajcic-Dragos V, Bougleux Gomes HA, Iyer V, et al. Three types of functional regulatory T cells control T cell responses at the human maternal-fetal Interface. Cell Reports. 2019;27(9):2537-2547.e5
  129. 129. Papuchova H, Kshirsagar S, Xu L, Bougleux Gomes HA, Li Q , Iyer V, et al. Three types of HLA-G+ extravillous trophoblasts that have distinct immune regulatory properties. Proceedings of the National Academy of Sciences of the United States of America. 2020;117(27):15772-15777
  130. 130. Hosseini Teshnizi S, Ali-Hassanzadeh M, Gharesi-Fard B, Kabelitz D, Kalantar K. Influence of forkhead box protein 3 polymorphisms (rs2232365, rs3761548) with the outcome of pregnancy: A meta-analysis. Journal Cellular Physiology. 2019;234(9):16573-16581
  131. 131. Tsuda S, Zhang X, Hamana H, Shima T, Ushijima A, Tsuda K, et al. Clonally expanded Decidual effector regulatory T cells increase in late gestation of Normal pregnancy, but not in preeclampsia, in humans. Frontiers in Immunology. 2018;9:1934
  132. 132. Vince GS, Starkey PM, Austgulen R, Kwiatkowski D, Redman CW. Interleukin-6, tumour necrosis factor and soluble tumour necrosis factor receptors in women with pre-eclampsia. British Journal of Obstetrics and Gynaecology. 1995;102(1):20-25
  133. 133. Schumacher A, Costa SD, Zenclussen AC. Endocrine factors modulating immune responses in pregnancy. Frontiers in Immunology. 2014 [cité 9 sept 2022];5:1-12. DOI: 10.3389/fimmu.2014.00196/abstract
  134. 134. Mizoguchi A, Mizoguchi E, Takedatsu H, Blumberg RS, Bhan AK. Chronic intestinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation. Immunity. 2002;16(2):219-230
  135. 135. Robertson SA, Care AS, Skinner RJ. Interleukin 10 regulates inflammatory cytokine synthesis to protect against lipopolysaccharide-induced abortion and fetal growth restriction in Mice1. Biology of Reproduction. 2007;76(5):738-748
  136. 136. Murphy SP, Fast LD, Hanna NN, Sharma S. Uterine NK cells mediate inflammation-induced fetal demise in IL-10-null mice. Journal of Immunology. 2005;175(6):4084-4090
  137. 137. White CA, Johansson M, Roberts CT, Ramsay AJ, Robertson SA. Effect of Interleukin-10 null mutation on maternal immune response and reproductive outcome in Mice1. Biology of Reproduction. 2004;70(1):123-131
  138. 138. Busse M, Redlich A, Hartig R, Costa SD, Rathert H, Fest S, et al. Imbalance between inflammatory and regulatory cord blood B cells following pre-term birth. Journal of Reproductive Immunology. 2021;145:103319
  139. 139. Guzman-Genuino RM, Hayball JD, Diener KR. Regulatory B cells: Dark horse in pregnancy immunotherapy? Journal of Molecular Biology. 2021;433(1):166596
  140. 140. Zenclussen AC. A novel mouse model for preeclampsia by transferring activated Th1 cells into Normal pregnant mice. In: Placenta and Trophoblast. New Jersey: Humana Press; 2005 [cité 9 sept 2022]. pp. 401-412. DOI: 10.1385/1-59259-989-3:401
  141. 141. Zenclussen AC, Fest S, Joachim R, Klapp BF, Arck PC. Introducing a mouse model for pre-eclampsia: Adoptive transfer of activated Th1 cells leads to pre-eclampsia-like symptoms exclusively in pregnant mice. European Journal of Immunology. 2004;34(2):377-387
  142. 142. Muzzio DO, Ziegler KB, Ehrhardt J, Zygmunt M, Jensen F. Marginal zone B cells emerge as a critical component of pregnancy well-being. Reproduction. 2016;151(1):29-37
  143. 143. Morandi F, Horenstein AL, Quarona V, Faini AC, Castella B, Srinivasan RC, et al. Ectonucleotidase expression on human amnion epithelial cells: Adenosinergic pathways and Dichotomic effects on immune effector cell populations. Journal of Immunology. 2019;202(3):724-735
  144. 144. Ribeiro A, Laranjeira P, Mendes S, Velada I, Leite C, Andrade P, et al. Mesenchymal stem cells from umbilical cord matrix, adipose tissue and bone marrow exhibit different capability to suppress peripheral blood B, natural killer and T cells. Stem Cell Research & Therapy. 2013;4(5):125
  145. 145. Ji YR, Yang ZX, Han ZB, Meng L, Liang L, Feng XM, et al. Mesenchymal stem cells support proliferation and terminal differentiation of B cells. Cellular Physiology and Biochemistry. 2012;30(6):1526-1537
  146. 146. Silini AR, Di Pietro R, Lang-Olip I, Alviano F, Banerjee A, Basile M, et al. Perinatal derivatives: Where do we stand? A roadmap of the human placenta and consensus for tissue and cell nomenclature. Frontiers in Bioengineering and Biotechnology. 2020;8:610544
  147. 147. Che N, Li X, Zhou S, Liu R, Shi D, Lu L, et al. Umbilical cord mesenchymal stem cells suppress B-cell proliferation and differentiation. Cellular Immunology. 2012;274(1-2):46-53
  148. 148. Lin HD, Fong CY, Biswas A, Choolani M, Bongso A. Human Wharton’s jelly stem cells, its conditioned medium and cell-free lysate inhibit the growth of human lymphoma cells. Stem Cell Reviews and Reports. 2014;10(4):573-586
  149. 149. Ma L, Zhou Z, Zhang D, Yang S, Wang J, Xue F, et al. Immunosuppressive function of mesenchymal stem cells from human umbilical cord matrix in immune thrombocytopenia patients. Thrombosis and Haemostasis. 2012;107(5):937-950
  150. 150. Xue Q , Yin Z, Varshithreddy N, Liang H s, Wang M y, Dong W l, et al. The immunomodulatory function of human amniotic fluid stromal cells on B lymphocytes. Journal of Neurorestoratology. 2018;6(1):122-133
  151. 151. Jensen F, Wallukat G, Herse F, Budner O, El-Mousleh T, Costa SD, et al. CD19+CD5+ cells as indicators of preeclampsia. Hypertension. 2012;59(4):861-868
  152. 152. Eledel R, Bassuoni M, Radwan W, Masoud A, Eldeeb S. CD19 + CD5 + B-cell expansion and risk of pre-eclampsia. Menoufia Medical Journal. 2016;29(2):319
  153. 153. Wallukat G, Homuth V, Fischer T, Lindschau C, Horstkamp B, Jüpner A, et al. Patients with preeclampsia develop agonistic autoantibodies against the angiotensin AT1 receptor. The Journal of Clinical Investigation. 1999;103(7):945-952
  154. 154. Silini AR, Masserdotti A, Papait A, Parolini O. Shaping the future of perinatal cells: Lessons from the past and interpretations of the present. Frontiers in Bioengineering and Biotechnology. 2019;7:75
  155. 155. Chen K, Wang D, Du WT, Han ZB, Ren H, Chi Y, et al. Human umbilical cord mesenchymal stem cells hUC-MSCs exert immunosuppressive activities through a PGE2-dependent mechanism. Clinical Immunology. 2010;135(3):448-458
  156. 156. Rossi D, Pianta S, Magatti M, Sedlmayr P, Parolini O. Characterization of the conditioned medium from amniotic membrane cells: Prostaglandins as key effectors of its immunomodulatory activity. PLoS One. 2012;7(10):e46956
  157. 157. Magatti M, Vertua E, De Munari S, Caro M, Caruso M, Silini A, et al. Human amnion favours tissue repair by inducing the M1-to-M2 switch and enhancing M2 macrophage features. Journal of Tissue Engineering and Regenerative Medicine. 2017;11(10):2895-2911
  158. 158. Cargnoni A, Ressel L, Rossi D, Poli A, Arienti D, Lombardi G, et al. Conditioned medium from amniotic mesenchymal tissue cells reduces progression of bleomycin-induced lung fibrosis. Cytotherapy. 2012;14(2):153-161
  159. 159. Carbone A, Castellani S, Favia M, Diana A, Paracchini V, Di Gioia S, et al. Correction of defective CFTR/ENaC function and tightness of cystic fibrosis airway epithelium by amniotic mesenchymal stromal (stem) cells. Journal of Cellular and Molecular Medicine. 2014;18(8):1631-1643
  160. 160. Lee PH, Tu CT, Hsiao CC, Tsai MS, Ho CM, Cheng NC, et al. Antifibrotic activity of human placental amnion membrane-derived CD34+ mesenchymal stem/progenitor cell transplantation in mice with Thioacetamide-induced liver injury. Stem Cells Translational Medicine. 2016;5(11):1473-1484
  161. 161. Parolini O, Souza-Moreira L, O’Valle F, Magatti M, Hernandez-Cortes P, Gonzalez-Rey E, et al. Therapeutic effect of human amniotic membrane-derived cells on experimental arthritis and other inflammatory disorders: Effects of HAMCs on experimental arthritis. Arthritis & Rheumatology. 2014;66(2):327-339
  162. 162. Tuca AC, Ertl J, Hingerl K, Pichlsberger M, Fuchs J, Wurzer P, et al. Comparison of Matrigel and Matriderm as a carrier for human amnion-derived mesenchymal stem cells in wound healing. Placenta. 2016;48:99-103
  163. 163. Pischiutta F, Brunelli L, Romele P, Silini A, Sammali E, Paracchini L, et al. Protection of brain injury by amniotic mesenchymal stromal cell-secreted metabolites. Critical Care Medicine. 2016;44(11):e1118-e1131
  164. 164. Lin YC, Ko TL, Shih YH, Lin MYA, Fu TW, Hsiao HS, et al. Human umbilical mesenchymal stem cells promote recovery after ischemic stroke. Stroke. 2011;42(7):2045-2053
  165. 165. Giampà C, Alvino A, Magatti M, Silini AR, Cardinale A, Paldino E, et al. Conditioned medium from amniotic cells protects striatal degeneration and ameliorates motor deficits in the R6/2 mouse model of Huntington’s disease. Journal of Cellular and Molecular Medicine. 2019;23(2):1581-1592
  166. 166. Wang H, Qiu X, Ni P, Qiu X, Lin X, Wu W, et al. Immunological characteristics of human umbilical cord mesenchymal stem cells and the therapeutic effects of their transplantion on hyperglycemia in diabetic rats. International Journal of Molecular Medicine. 2014;33(2):263-270
  167. 167. Tan B, Yuan W, Li J, Yang P, Ge Z, Liu J, et al. Therapeutic effect of human amniotic epithelial cells in murine models of Hashimoto’s thyroiditis and systemic lupus erythematosus. Cytotherapy. 2018;20(10):1247-1258
  168. 168. Gao L, Zhang Y, Hu B, Liu J, Kong P, Lou S, et al. Phase II multicenter, randomized, double-blind controlled study of efficacy and safety of umbilical cord-derived mesenchymal stromal cells in the prophylaxis of chronic graft-versus-host disease after HLA-Haploidentical stem-cell transplantation. Journal of Clinical Oncology. 2016;34(24):2843-2850
  169. 169. Papait A, Vertua E, Magatti M, Ceccariglia S, De Munari S, Silini AR, et al. Mesenchymal stromal cells from fetal and maternal placenta possess key similarities and differences: Potential implications for their applications in regenerative medicine. Cell. 2020;9(1):E127

Written By

Gatien A.G. Lokossou and Maximin Senou

Submitted: 12 September 2022 Reviewed: 28 September 2022 Published: 18 January 2023