Abstract
Macrophages (MФs) are the leukocytes produced from differentiation of monocytes and are located in almost all tissues of human body. They are involved in various processes, such as phagocytosis, innate and adaptive immunity, proinflammatory (M1) and anti-inflammatory (M2) activity, depending on the tissue microenvironment. They play a crucial role in pregnancy, and their dysfunction or alteration of polarity is involved in pregnancy disorders, like preeclampsia, recurrent spontaneous abortion, infertility, intrauterine growth restriction, and preterm labor. About 50–60% of decidual leukocytes are natural killer (NK) cells followed by MФs (the second largest population). MФs are actively involved in trophoblast invasion, tissue and vascular remodeling during early pregnancy, besides their role as major antigen-presenting cells in the decidua. These cells have different phenotypes and polarities in different stages of pregnancy. They have also been observed to enhance tumor growth by their anti-inflammatory activity (M2 type) and prevent immunogenic rejection. Targeted alteration of polarity (M1–M2 or vice versa) could be a major focus in the future treatment of pregnancy complications. This review is focused on the role of MФs in pregnancy, their involvement in pregnancy disorders, and decidual MФs as possible therapeutic targets for the treatment of pregnancy complications.
Keywords: Macrophage, Human pregnancy, Decidualization, Polarity, VEGF, Preeclampsia
Introduction
The trophoblast cells of the implanted blastocyst invade the uterine endometrium of the mother through a process called placentation. The endometrial stromal cells differentiate (decidualization) and establish an environment conducive to trophoblast invasion (Hunt 2006). Three types of placentation exist among mammals—epitheliochorial, endotheliochorial, and hemochorial placentation (Vogel 2005) based on the contact pattern of trophoblast cells and the uterine lining. Hemochorial placentation is seen in human and mice, where the fetal membrane directly contacts with the maternal tissue and blood. This deep placentation signifies appropriate recognition and tolerance to the semiallogeneic fetus for a successful pregnancy, where maternal immune cells play a critical role (Wildman et al. 2006).
The leukocyte population in the uterus undergoes significant changes after implantation and decidual development. The T-cell and B-cell aggregates observed in the cycling uterus disappear, along with intangible eosinophils and mast cells. Natural killer (NK) cells and macrophages (MФs) dominate during early pregnancy (Hunt et al. 2000). Large number of leukocytes (MФs, NK cells, T cells, and dendritic cells) infiltrate into the pregnant decidua. Almost 40% of all cells in the decidua in the first trimester are leukocytes (von Rango 2008). An estimated 20–30% of leukocytes are MФs, playing a role in tolerance to the semiallogeneic fetus, trophoblast invasion, and tissue and vascular remodeling (Mizuno et al. 1994; Lidström et al. 2003; Nagamatsu and Schust 2010a; Renaud and Graham 2008; Li et al. 2019). The factors that attract MФs to the decidua include chemokines, colony-stimulating factor-1 (CSF-1) and granulocyte macrophage-colony-stimulating factor (GM-CSF), which are mostly produced in response to hormonal stimulation (Pollard 1998). The vascular endothelial growth factor receptor-1 (VEGFR-1) plays a pivotal role in recruitment of MФs and angiogenesis at implantation sites (Douglas et al. 2014). MФs can be distinguished from other leukocytes by distinct cell surface markers, such as CD14, CD68, and human leukocyte antigen (HLA) like HLA-DR (Heikkinen et al. 2003; Nagamatsu and Schust 2010a). They have plasticity to alter their functions in response to a changing microenvironment in tissues, thus widely involved in physiological functions and diseases (Stout and Suttles 2004). They are classified as classically activated (M1) and alternatively activated (M2) phenotypes based on their function and repertoire of cytokine production (Gordon 2003; Martinez et al. 2008) (Fig. 1). The M1MФs are proinflammatory in nature and are induced by pathogen (lipopolysaccharide) exposure, and tissue damage through interferon-γ (IFN-γ) and tumor necrosis factor (TNF) proteins (Martinez et al. 2008; Mosser 2003). These MФs produce reactive oxygen species (ROS) and nitric oxide (NO) from arginine by upregulation of the enzyme inducible NO Synthase (iNOS), to kill pathogens. They also produce higher level of cytokines like IL-12 and IL-23, and lower IL-10 level (Verreck et al. 2004). M2MФs are induced by Th2 cytokines, such as IL-4, and IL-13, as well as by anti-inflammatory IL-10, apoptotic cells, and macrophage colony-stimulating factor (MCSF, also called CSF-1) (Mantovani et al. 2004; Gough et al. 2001). These MФs produce higher level of IL-10 and lower level of IL-12 and IL-23 cytokines (Mills et al. 2000). They produce ornithine from arginine by upregulating arginase level and are anti-inflammatory in nature, involved in scavenging the apoptotic cells, tissue repair and remodeling (Xu et al. 2006). MФs predominately expressing iNOS over arginase are termed M1MФs and those predominately expressing arginase over iNOS are termed M2MФs. Studies reveal that decidual MФs acquire an M2 phenotype, promoting immune tolerance toward the fetus, required for successful pregnancy (Lidström et al. 2003). Recent study (Zhang et al. 2019) has revealed that the programmed cell death-1 (PD-1)/PD-1 ligand-1 (PDL-1) signaling regulates the macrophage differentiation in early pregnancy, promoting the polarization to M2 phenotype which supports the pregnancy. In fact, tissue MФs acquire different functional phenotypes ranging from proinflammatory (M1) to anti-inflammatory (M2) in response to the environmental milieu. It has been observed that M1MФs skew to the M2 phenotype when cultured in 1st trimester gravid serum, as revealed by a decrease in procalcitonin (a marker for evaluating bacterial infections) level, indicative of the M2 type (Rami et al. 2014). However, the M1/M2 concept has been reviewed, and it has been found that decidual MФs do not follow the typical M1/M2 paradigm, and that the M2 phenotype is not always induced by Th2 cytokines. Rather M-CSF and IL-10 are found to be efficient M2MФ markers’ (CD163 and CD206) inducers (Houser et al. 2011; Svensson et al. 2011). Another classification system separates MФs into classically activated MФs (CAMs) or type-I MФs, and alternatively activated MФs (AAMs) or type-II MФs (Porcheray et al. 2005). Most of the tumor-associated MФs (TAMs) are type-II (Tripathi et al. 2014). Gene expression analysis of decidual MФs has revealed an AAM phenotype (Gustafsson et al. 2008). There are few reviews discussing the role of decidual MФs in different stages of pregnancy (Ning et al. 2016; Svensson-Arvelund and Ernerudh 2015; Zhang et al. 2017). In this review, we have discussed the detailed role of MФs in pregnancy, their involvement in pregnancy disorders, and the possibility of these cells as therapeutic targets.
Fig. 1.

Macrophage differentiation and polarization in the decidua during pregnancy
Current Paradigms of MФ Differentiation and Function
Several hypotheses exist pertaining to the origin of endometrial leukocytes (Lee et al. 2015). One hypothesis describes leukocytes coming to endometrium through circulating blood. In this process, the circulating leukocytes extrava-sated from blood vessels across endothelial cell barriers. Endothelial cells express adhesion molecules which facilitate invasion by binding as ligands to homing receptors of immune cells (Johansson et al. 1999). Another hypothesis describes in situ proliferation of the resident immune cells (van den Heuvel et al. 2005; Kitaya et al. 2007). Yet another hypothesis proposes that hematopoietic precursors are recruited to and differentiate within the endometrium (Keskin et al. 2007; Manaster and Mandelboim 2010). Immune cells recruited from circulation probably undergo tissue-specific differentiation in response to local factors, and conferring characteristics on these cells that deviate from those of the parental cells (Keskin et al. 2007; King 2000).
About 50–60% of decidual leukocytes are NK cells followed by MФs (Houser 2012; Faas and de Vos 2017). Regulatory molecules such as steroid hormones, growth factors, bioactive peptides, and lipids influence the activities of these multifunctional cells in pregnant uterus and placenta, thus contributing to the complex cellular and molecular interactions required for successful pregnancy. The circulating blood monocytes are influenced by multiple factors to migrate and differentiate into tissue MФs, which express tissue-specific subpopulation markers (Gordon 2003). Movement of blood monocytes to tissue depends on specific cytokines and chemokines and their receptors. Tissue-specific environmental signals induce specific phenotypic profiles (McIntire et al. 2008). The decidual MФs of mammals have drawn remarkable attention for their wide variety of functions, such as (a) phagocytosis and tissue remodeling, (b) defense against infections, (c) influence over the function of surrounding cells like invasive cytotrophoblasts, glandular epithelium, and endothelial cells of arteries, (d) antigen presentation, and (e) production of cytokines, such as IL-10 and IL-15, which leads to successful pregnancy (Laskarin et al. 2005; Lidström et al. 2003; Heikkinen et al. 2003; Mor and Abrahams 2003; Reister et al. 2001; Verma et al. 2000). Their recruitment and function are influenced by conditioning factors in the human uterus. The protein IFN-γ, produced by NK cells, activates MФs in the pregnant uterus of mice and humans, leading to display of HLA class II antigens (Xie et al. 2005). The protein CSF-I (M-CSF) is another crucial factor, contributing to the development of the phenotypic profile of MФs in the pregnant uterus. This protein is highly expressed by the uterine epithelial cells of pregnant mice and humans (Pollard 1997) Besides, the decidual stromal cells are also important source for M-CSF production.
MФs in the Female Reproductive Tract
There is a strong impact on fertility and reproductive health from the interaction between immune system and female reproductive system. Physiological events in the female reproductive system, such as ovulation, menstruation, implantation, and onset of labor, are inflammatory processes. MФs play a crucial role in the initiation and resolution of inflammation, and they are detected in all phases of the menstrual cycle, scattered throughout the endometrium, being more prevalent around the endometrial gland (Yang et al. 2011; Salamonsen and Lathbury 2000). Their population increases in the early secretory phase and continues to increase in the late secretory phase of the menstrual cycle, with the sex hormones (estrogen and progesterone) playing a major role in their distribution in the endometrium (DeLoia et al. 2002; Keenihan and Robertson 2004; Jones et al. 2004). As uterine CD45+ leukocytes do not express estrogen and progesterone receptors, these sex hormones are likely to control uterine leukocytes indirectly. There is a cross-talk between the decidual immune cells, endometrial cells, epithelial cells, and stromal cells, delicately controlled by estrogen and progesterone (Salamonsen and Lathbury 2000). Physiologic levels of estrogen induce MФ proliferation and function, and estrogen has both positive and negative regulatory effects on the production of the chemokine CCL2, an MФ chemoattractant (Carruba et al. 2003). Besides CCL2, the level of other MФ chemoattractants, such as CCL7, CX3CL1, and CCL4, also increases in the perimenstrual period in the endometrium (Keenihan and Robertson 2004; Jones et al. 2004; Wira et al. 2005). Vaginal MФ number remains constant during the menstrual cycle (Amjadi et al. 2014). MФs have been observed to modulate ovarian functions, such as follicular atresia and luteolysis (Pate and Landis Keyes 2001; Penny 2000; Duncan 2000). The theca cell layers of the ovary are rich in these cells, where matrix metalloproteinases (MMPs) (secreted by MФs) get activated during ovulation. Endometrial MФs have been observed to have an M2-like phenotype (CD68+CD163+CD206+IL10high) like those of the decidua; however, they express certain markers differently and secrete different amounts of pro- and anti-inflammatory cytokines and, hence, belong to different subpopulations (Quillay et al. 2015). Endometrial MФs express CD163, CD80, CD64, CD16, CD11c, CCR5, CXCR4, and DC-SIGN significantly less strongly than do decidual MФs. Furthermore, decidual MФs play a role in the phagocytosis of apoptotic trophoblasts, whereas endometrial MФs phagocytose endometrial tissue debris (Thiruchelvam et al. 2013).
MФs in Fertilization, Implantation, and Decidualization
The mid-secretory phase (days 19–23 of the menstrual cycle) is called the ‘window of implantation’, when the uterus becomes receptive (Gnainsky et al. 2010). There is an increased level of proinflammatory cytokines, such as IL-6, leukemia inhibitory factor (LIF), and TNF-α in the endometrium which characterize early implantation (van Mourik et al. 2009).
MФs and dendritic cells (DCs) play a crucial role in decidualization and implantation (Lea and Clark 1991; Gardner and Moffett 2003; Plaks et al. 2008; Blois et al. 2004). They are involved in tissue remodeling and angiogenesis through the production of an array of cytokines, chemokines, and enzymes (Abrahams et al. 2004; David Dong et al. 2009). Additionally, these molecules target the luminal epithelium and subsequently contribute to the acquisition of endometrial receptivity. It has been proposed that the increased receptivity of the uterus after biopsy treatment may be mediated by the immune cells recruited by the chemoattractant molecule macrophage inflammatory protein-1β (MIP-1-β, also known as CCL4) (Gnainsky et al. 2010). The regulatory protein osteopontin, which is an adhesive molecule and a biomarker of receptive endometrium, seems to regulate MIP-1-β secretion positively, facilitating the recruitment of MФs/DCs (Goetzl et al. 1996).
Several regulatory proteins play a vital role in implantation of the embryo. The level of the protein CSF-I (secreted by uterine epithelium) increases at the time of implantation and reaches its peak during placentation (Horcajadas et al. 2007). DCs secrete sFlt-1(Flt-1: Fms-like tyrosine kinase 1; also called VEGFR1) and transforming growth factor-β1 (TGF-β1) which regulate endometrial angiogenesis and blood vessel maturation and are also involved in Tregulatory cell (Treg cell) development (Gardner and Moffett 2003). M2 polarization process (induced by glucocorticoids and the Th2 cytokines) enhances innate immunity receptors, such as scavenger receptors and MФ mannose receptors, part of the M2 phenotype, which supports implantation (Nagamatsu and Schust 2010b; Pollard 2008; Laskarin et al. 2005).
Additionally, the transcription factor PU.1 (Ets–family member) is observed to regulate the development and differentiation of MФs, B cells, T cells, and NK cells (Lee et al. 2011; Dakic et al. 2005). This transcription factor also modulates the C1q (the first component of classical component activation) gene expression in MФs and DCs (Iwasaki et al. 2005). The C1q component is significantly involved in fetal tolerance, migration of trophoblasts, and spiral artery remodeling. Thus, PU.1 plays a crucial role in implantation through regulation of C1q expression in decidua (Chen et al. 2011).
Tissue MФs influence the biochemical events occurring in the endometrial cell lining in early pregnancy to promote uterine receptivity and facilitate embryo attachment and implantation. In this process, LIF (derived from MФs) regulates the structure of surface glycans in epithelial cells and makes the endometrium receptive (Madhukaran et al. 2015). Cell–cell communication is very crucial for maternal–embryonic recognition, and endometrial MФs act as determinants of uterine receptivity. It has been observed that the two dendritic cell-associated molecules CD200 and MD-1 have immune regulatory activity toward MФs, contributing to successful pregnancy. Increased expression of CD200R in MФs enhances indoleamine 2,3-dioxygenase (IDO) activity, thus contributing to an immune-suppressive environment needed for successful implantation (Nakamura et al. 2012).
Regulation and Function of MФs at the Maternal–Fetal Interface
MФs play an important role in the preparation of a receptive endometrium during the window of implantation and subsequently decidualization of the endometrial stroma (Gorczynski et al. 2002).
MФs in Early Pregnancy
Decidual MФs play vital roles in apoptosis and cell clearance during pregnancy. The quick and effective removal of apoptotic cells prevents the leakage of self-antigens and inflammation. MФs have a capacity for alloantigen presentation, immunosuppressive activity and less ability to produce IL-1 than do peripheral monocytes (Mizuno et al. 1994). Their immune-suppressive activity may be due to their high levels of IL-10 and IDO production, along with the production of prostaglandin-E2 (PGE2), which blocks the activation of cytotoxic leukocytes (Kats et al. 2005). IDO degrades tryptophan and thereby prevents maternal T cell activation, which requires tryptophan. It has been observed that serum tryptophan levels decrease from the first trimester of human pregnancy (Miwa et al. 2005). IDO expression in MФs is regulated by CD4+ CD25+ Treg cells, which upregulate their IDO levels through cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), present on the cell surface (Schrocksnadel et al. 1996) (Fig. 2). The enzyme IDO is also secreted by decidual DCs, extravillous trophoblasts (EVTs), and villous trophoblasts in human (Sedlmayr et al. 2002; Kudo et al. 2004; Honig et al. 2004; Hwu et al. 2000; Munn et al. 2002). Many MФ-derived proangiogenic factors are involved in vascular remodeling for proper uteroplacental circulation. MФs and NK cells are enriched in the vicinity of the trophoblast invasion front during early pregnancy (Helige et al. 2014). Both cell types are involved in spiral arteriole remodeling. The vascular remodeling process occurs in distinct trophoblast-independent (early changes) and trophoblast-dependent stages. In the early stage, vascular smooth muscle cells are disrupted and disorganized before the arrival of endovascular trophoblasts. This process is associated with MФ and NK cell infiltration, secretion of MMP-7 and −9, and apoptosis of smooth muscle cells and endothelial cells (Smith et al. 2009). When EVTs invade the decidua, they come across NK cells and MФs (NK cells and MФs are in close proximity) and cross-talk among those cells occur within the uterine wall. It has been found that NK cells have a cytolytic activity toward EVTs, but this activity is prevented by MФs through a TGF-β1-dependent mechanism (Co et al. 2013). EVTs are unique in their expression of HLA molecules, as they do not have the highly polymorphic (HLA-A and HLA-B) classical class I HLAs, but they express HLA-C and the non-classical (HLA-E, HLA-F, and HLA–G) class-I molecules (King et al. 2000a, b; Shobu et al. 2006). The HLA-G molecule has four membrane-bound (HLA-G1–4) and three soluble isoforms (HLA-G5–7) produced by alternative splicing (Shakhawat et al. 2010). Decidual MФs express the HLA-G receptors leukocyte Ig-like receptor B1 (LILRB1) (also known as Ig-like transcript-2, ILT2) and LILRB2 (ILT4). Trophoblast-derived HLA molecules HLA-C and HLA-G are observed to regulate MФ function by making them immune tolerant through interaction with the ILT2 and ILT4 receptors (Petroff et al. 2002). The HLA-G homodimer engagement of ILT2 on MФs triggers the secretion of IL-6, IL-8, and to a lesser degree TNF-α (Li et al. 2009). In addition, HLA-G5 (soluble form) induces IL-4 secretion, which is important for successful pregnancy, acting to modulate expression of the ILT2 receptor on TH cells and MФs (Lombardelli et al. 2013).
Fig. 2.

Treg cells showing immunotolerance through M2 MФs and dendritic cells (DCs). The naïve T cells are induced by IL-2 and TGF-β in the decidua, forming Treg cells. Treg cells express the surface molecule CTLA4 which interacts with CD80/CD86 of DCs and MФs, thus inducing those cells for IDO production. IDO inhibits Teff cell proliferation. The cytokines IL-10, IL-35, and TGF-β produced from Treg cells also inhibit Teff cell proliferation. IDO = Indoleamine 2, 3-dioxygenase, Treg = T regulatory, Teff = T effector
Costimulatory molecules play a significant role in immu-nomodulation by regulating signaling pathways. The B7 family is a major costimulatory ligand group that includes B7.1, B7.2, B7-H1, B7-DC, B7-H2, B7-H3, B7-H4, and B7-H6 (Sayama et al. 2013). MФs of early pregnancy express the B7-H1 ligand molecule, which is inhibitory in action. Expression of this ligand molecule is not observed in term MΦs and peripheral monocytes of pregnant women. This ligand binds to its receptor programmed death-1 (PD-1), which is highly expressed in T cells, and this interaction suppresses IFN-γ production by activated T cells, thus making them immune suppressive. This inhibitory signaling fine tunes the IFN-γ level and balances the maternal immune response during early human pregnancy (Sayama et al. 2013). There is also downregulation of PI3/Akt pathway signaling leading to inhibition of the T cell activity (Parry et al. 2005). The MФ-derived cytokine IL-33 is found to be a critical factor for placental growth in early pregnancy, increasing the proliferation of primary trophoblasts, villous cytotrophoblasts, and cell column trophoblasts. IL-33 is a member of the IL-1 family, and MФs seem to maintain its expression until term. IL-33-induced proliferation is mediated by PI3 K/AKT and MAPK/ERK signaling (Fock et al. 2013). It has also been observed that GM-CSF and M-CSF, secreted from induced first trimester decidual cells, promote MФ activation, and these activated cells induce EVT apoptosis through a caspase 3/7-dependent pathway (Wu et al. 2012).
MФs in Late Pregnancy
MФ number decreases at term, becoming the smallest component of decidual leukocytes, whereas CD3+ T lymphocytes are the largest (Williams et al. 2009). However, their number increases by infiltration during term and preterm labor in human and rat decidua, showing evidence of decidual inflammation preceding labor (Hamilton et al. 2012). MФs accumulated in the lower uterine segment are involved in cervical ripening during late pregnancy. They play prominent roles in intrauterine defense by producing superoxide radicals and TNF-α in response to pathogens, thus protecting the fetus against infections and preventing preterm labor (Singh et al. 2005). There is alteration in the level of maternal immune tolerance toward fetal antigens at the initiation of labor. The subpopulation of FoxP3+ T cells (Treg cells) seems to decrease, whereas the immune-suppressive B7-H4+ MФ subpopulation remains unchanged as labor progresses. Besides, IL-10 produced by the decidual MФs contributes to the immune-suppressive environment (Heikkinen et al. 2003). This phenomenon reflects a balancing act of brief activation of the maternal immune system with an associated mechanism to restrict this action for term delivery (Galazka et al. 2009). Preterm delivery (PTD) and term delivery probably follow a common downstream pathway with increased release of MMP and collagen degradation; however, the source of MMP seems to be MФs in PTD, and cervical stromal fibroblasts and columnar epithelial cells at term. The trigger mechanism also seems to be different, with progesterone withdrawal triggering MMP release at term and anaphylatoxin C5a as a trigger for MMP release in PTD (Gonzalez et al. 2011).
A multi-domain protein stabilin-1, present in lymphatic and vascular endothelial cells and in type-II MФs, acts as a scavenging receptor and adhesion molecule. Analysis of its expression on MФs of term placenta has revealed its presence both in decidual and placental MФs; these may be involved in regulating leukocyte migration and scavenging during fetal development (Palani et al. 2011). In addition, the systemic hormone relaxin is secreted from the corpus luteum (CL) at low level during pregnancy and high level in late pregnancy and early parturition (Lafayette et al. 2011). This hormone modulates the activity of MФs by acting on both the glucocorticoid receptor (GR) and relaxin receptor expressed on these cells. The low level of relaxin during pregnancy may be anti-inflammatory, acting through the GR and supporting pregnancy, whereas its high level acts through the relaxin receptor, increasing proinflammatory IL-6 production by MФs, which contributes to a sterile inflammatory milieu required for initiation of parturition (Horton et al. 2011).
Angiogenic Functions of Decidual MФs
Angiogenesis (development of blood vessels from preexisting vasculature) plays a crucial role in both normal and pathological conditions, and the VEGFR isoforms VEGFR-1, −2, and −3 play a key role as mediators of angiogenic response (Petrova et al. 1999). Of these isoforms, VEGFR-1 plays an important role in angiogenesis and MФ recruitment at implantation sites. The role of VEGF (paracrine signal) in making and maintaining blood vessels has been studied comprehensively (Heloterä and Alitalo 2007). Vascular homeostasis is maintained by VEGF produced endogenously by endothelial cells (Lee et al. 2007).
Decidual MФs act significantly in angiogenesis and spiral artery remodeling during the first and second trimester, establishing maternal blood flow essential for the growth of the fetus. During these processes, there is a remarkable loss of vascular smooth muscle, replacement of endothelial cells by EVTs, and decreased resistance by the spiral arteries to blood flow to the placenta (Burrows et al. 1996). MФs produce various proangiogenic factors in response to hypoxia and other micro-environmental signals as observed in tumor progression and wound healing (Coffelt et al. 2009). Similar hypoxic conditions also prevail in the decidual endometrium during the first trimester. There is an abundant infiltration of MФs and NK cells in the initial stage of vascular remodeling and these leukocytes produce MMP-9 and −7, which degrade the ECM and promote endovascular trophoblast invasion. There is impaired vascular development in conditionally ablated CD11c+ DCs during implantation in transgenic mice, demonstrating the importance of MФs in fine-tuning uteroplacental circulation in human pregnancy (Gardner and Moffett 2003).
The VEGF family members play a crucial role in angiogenesis and vasculature development in pregnancy (Sugino et al. 2002; Zhou et al. 2002). The VEGF protein acts by binding the receptors Flt-1 and kinase insert domain receptor (KDR). Another member of the VEGF family, placental growth factor (PlGF), is produced by the placenta and acts by binding to Flt-1 (Clark et al. 1998). VEGF and Flt-1 expression are increased in MФs during early pregnancy, regulated by hormones. EVTs also produce VEGF and PlGF abundantly along with the MФs. PlGF causes activation of monocytes via activation of Flt-1 (Selvaraj et al. 2003). Flt-1 is observed as a functional receptor for VEGF and PlGF in monocytes and endothelial cells, and acts as a mediator of monocyte recruitment (Clauss et al. 1996). Studies in our laboratory have shown that VEGF may play a crucial role in MФ recruitment and M2 polarization in the human decidua (Wheeler et al. 2017).
Phenotypes of Decidual MФs
Immature monocytes are released from the bone marrow, recruited to tissues by chemokines after circulating in the blood, and differentiate into MФs (Imhof and Aurrand-Lions 2004). Few studies have been performed on phenotyping MФ lineages and subsets. MФs possess functional plasticity with reversible adaptation to changing environments. The ability of monocytes and tissue MФs to adapt to variation in their microenvironment challenges the assumption that MФs display unique specific markers and makes it difficult to categorize them in particular lineages.
MФ polarization has been observed between M1 and M2 phenotypes throughout pregnancy (Table 1). During the peri-implantation period, the M1 type predominates, after which there is a transition to a mixed population of M1 and M2 types, during which trophoblasts invade the stroma and settle in the endometrial lining (Jaiswal et al. 2012). The mixed population thrives until early in the second trimester (Mor et al. 2011). Subsequently, the pro-M2 phenotype predominates as the placenta is established, and this prevents rejection of the fetus. It is likely that the immune-suppressive environment is maintained throughout pregnancy by the M2 macrophages. These phenotypes dominate in the decidua, and they are responsible for immunotolerance, which is a prerequisite for successful implantation and fetal development. In fact, the polarization of M1 phenotype to M2 phenotype during second trimester and again back to M1 phenotype at delivery is not well defined and needs further study.
Table 1.
Different phenotypes of macrophages during pregnancy
| MФ phenotype | Cytokine production | Function | References |
|---|---|---|---|
| Ml | IL-12, IL-1β, TNF-α, IL-6, NO, CXCL10, CCR7 | Promotes embryo implantation | Wheeler et al. (2017), Zhang et al. (2017) |
| M2a | Fibronectin, Arginase-1, IL-10, IL-6, CCL-17, CCL-18 | Th2 activation, tissue repair | Wheeler et al. (2017), Ning et al. (2016) |
| M2b | IL-10, IL-1β, IL-12low | Immunoregulatory and Th2 activation | Ning et al. (2016) |
| M2c | IL-10, TGF-β | Immunotolerance, matrix remodeling and repair | Ning et al. (2016) |
| M2d | IL-10, VEGF | Proangiogenic | Ning et al. (2016) |
| CD14 + CD11CHI | Upregulates CD1, genes associated with lipid metabolism, inflammation, Ag presentation | Antigen processing and presentation | Houser (2012) |
| CD14 + CD11CLO | Expresses genes associated with ECM formation, tissue growth, etc. | Homeostatic function during placental growth | Houser (2012) |
CD14−/CD68+ cells were found to be more abundant in the decidua and CD14+/CD68+ cells more abundant in the myometrium of preeclampsia and preterm labor cases (Kim et al. 2007). A comparison of the MФ phenotypes in preterm preeclamptic, preterm control, and term control placentas showed higher expression of CD14 and CD163 in decidua basalis of preterm preeclamptic than that of preterm control cases (Schonkeren et al. 2011). The percentage of cells expressing CD163 (scavenger receptor), CD206 (mannose receptor), CD209 (DC-SIGN), and neuropilin-1 (NRP1) was found to be significantly higher in the CD14+ decidual MФ population than in CD14+ blood monocytes (Svensson et al. 2011). Comparison of CD14+ MФs and CD14+ monocytes revealed higher HLA-DR and lower CD86 (costimulatory) expression in MФs than in monocytes during pregnancy (Heikkinen et al. 2003). Some discrepancies were observed in the 1st trimester MФ phenotypes, with one study showing dominance of the M2 phenotype and the other study showing two distinct subsets of CD14+ decidual MФs (CD11CHI and CD11cLO) in 1st trimester decidual tissue in humans, not fitting the conventional M1/M2 categorization (Cupurdija et al. 2004; Houser et al. 2011; Houser 2012). A study on MФs of 1st trimester human deciduas showed that they express markers associated with alternative activation (M2). The markers stabilin-1 (MS-1) and coagulation factor XIIIa were present in the interior of decidual MФs (Cupurdija et al. 2004). These discrepancies may be attributed to the anatomical plane of the feto-maternal junction analyzed and varying antibodies used for detection (Kim et al. 2007).
MФ Dysfunction in Common Pregnancy‑Related Disorders
Infertility
MФs exhibit wide plasticity and are sensitive to small changes in the microenvironment. Factors like nutritional status, smoking habits, toxins (environmental), inflammatory conditions, and physiological stress impart adverse effects on fertility (Zhang and Mosser 2008; Martinez et al. 2009). Although endometrial MФ numbers do not correlate with infertility diagnoses, their phenotypes and cytokine secretory profiles have discernible effects (Dechaud et al. 1998; Leung et al. 1998). The colocalization of CD68 and the secreted cytokines IL1A, IL1B, and IL-6 show variation in conjunction with fertility status. Oviductal MФs are potential mediators of infertility and have been observed to have morphologic characteristics similar to those of peritoneal MФs (ability to phagocytose normal sperm) and occur in patients with infertility problems. These MФs might obstruct fertilization by phagocytosis of sperm (Haney et al. 1983).
Recurrent spontaneous abortion (RSA)
The protein cathepsin E is an endolysosomal aspartic proteinase expressed mainly in immune cells and is highly secreted by activated phagocytes (Nishioku et al. 2002). It prevents tumor growth and metastasis by inducing apoptosis, inhibiting angiogenesis, and increasing immune responses (Shin et al. 2007). A study on the role of cathepsin E in early pregnancy in decidual MФs of patients with recurrent miscarriage (RM) revealed decreased activity of cathepsin E produced by MФs, which may be responsible for induction of miscarriage (Goto et al. 2014). Studies have also shown the dysregulation of MФ activation by Treg cells, leading to unexplained RM cases (Wang et al. 2011). Thrombospondin 1 (TSP1) is mainly expressed in blood platelets which exists in tissue fluid in a free state but can bind the surfaces of MФs. The binding partners of TSP1 are CD36 (scavenger receptor), CD47, and heparin sulphate proteoglycan, which are expressed on MФs (Bornstein 1995; Yamauchi et al. 2002). TSP1 may play a role in regulating MФ function through interactions with these binding partners. A study on the effects of TSP1 on decidual MФs in RSA revealed that its expression was decreased in those cells, along with decreased IL-10 level, as compared to that of normal pregnant women. TSP1 also enhanced IL-10 expression in decidual MФs in vitro, suggesting its involvement in regulating secretion of IL-10 and induction of immune tolerance at the maternal–fetal interface (Jin et al. 2009).
Preeclampsia and intra‑uterine growth restriction (IUGR)
Decidual MФs induce apoptosis and limit endovascular trophoblast invasion in preeclamptic women. The uteroplacental arteries are invaded by trophoblasts and are largely devoid of MФs in normal pregnancy, whereas reduced trophoblast invasion of uteroplacental arteries and accumulations of apoptotic trophoblasts in the vicinity of arterial walls are found in IUGR and preeclampsia (Reister et al. 1999). An increase in trophoblast apoptosis may initiate inflammatory events, promoting further death of trophoblast cells and thus preventing normal trophoblast invasion, which may lead to pregnancy disorders, such as preeclampsia and IUGR (Mor and Abrahams 2003). The proinflammatory cytokines secreted by M1MФs increase Fas expression and enhance trophoblast sensitivity to Fas-mediated apoptosis (Aschkenazi et al. 2002; Neale et al. 2003). Hence, MФ-induced apoptosis affects the endovascular pathway of trophoblasts in pregnancy complications (Huppertz et al. 2006). Recent study (Li et al. 2019) revealed polarization of proinflammatory M1 subtype to anti-inflammatory M2 subtype through activation of Tim-3/Gal-9 signaling pathway in the preeclamptic rat model. There is a significant increase in MФ numbers in the decidua of preeclamptic patients (Lockwood et al. 2006). In contrast, Burk et al. (2001) observed reduced numbers of MФs with a CD14-, HLA-DR-, and mannose receptor-positive phenotype in placentas of preeclamptic women as compared to normal placentas (Burk et al. 2001). Pregnancies complicated by preeclampsia or IUGR showed activated MФs producing proinflammatory cytokines, such as TNF-α and IFN-γ, and inducing the apoptosis of EVTs (Blois et al. 2004).
Preeclampsia is accompanied by an increased number of both MФs and DCs at implantation sites, along with enhanced levels of recruiting chemokines, implicating impaired trophoblast invasion. There is an enhanced level of GM-CSF (a potent inducer of macrophage differentiation) in preeclamptic deciduas induced by the proinflammatory cytokines TNF-α and IL-1β, suggesting involvement of GM-CSF in the pathogenesis of preeclampsia (Huang et al. 2010). Hypoxic conditions, oxidative stress, and inflammation induce necrosis or aponecrosis of trophoblasts. MФs and DCs produce type-I cytokines, such as TNF-α, IL-12, and IFN-γ, after phagocytosis of necrotic and aponecrotic trophoblasts and aggravate the inflammation (Huppertz et al. 2003). This condition probably induces apoptosis of EVTs, culminating in poor placentation, as observed in preeclampsia (Saito and Sakai 2003). The hormone corticotropin-releasing hormone (CRH) plays a vital role in mediating the stress response through the activation of the hypothalamic–pituitary–adrenal axis (Bale and Vale 2004). Evidence suggests a role for CRH in reproductive physiology, with its abnormal function implicated in the pathogenesis of preeclampsia (Karteris et al. 2005). CRH and its receptors are expressed in the placenta and are involved in regulation of trophoblast invasion, placental vasculature, myometrial contractility, and onset of labor. CRH expression is upregulated in preeclamptic placentas, inducing FasL expression in maternal MФs. These activated MФs cause Fas-mediated EVT apoptosis, perturbing placentation (Petsas et al. 2012).
Preterm Labor
It is important to understand the host immune responses against uterine invading microbes in early pregnancy, as there is a correlation persisting between extremely premature births and intrauterine infection. It has been observed that MФs modulate the decidual immune response against Listeria monocytogenes infections in early pregnancy in mice (Qiu et al. 2009). The MФ migration inhibitory factor (MIF) is a soluble proinflammatory cytokine released by activated leukocytes (Bevilacqua et al. 2014). MIF inhibits random MФ migration from peritoneal exudates, and its expression is confined to proliferative villous cytotrophoblasts and extravillous cell columns, indicating regulatory roles in the process of embryo implantation and pregnancy maintenance (Bucala 2007; Calandra and Roger 2003; Arcuri et al. 1999, 2007). Decreased maternal MIF serum level is linked with recurrent miscarriage in early gestation, whereas increased levels in early and mid-gestation are associated with subsequent preterm delivery (Yamada et al. 2003; Pearce et al. 2008, 2010). Enhanced maternal serum levels of MIF and altered expression of other proteins in placental compartments is also observed in preeclampsia, implicating MIF as a potential target for therapy (Todros et al. 2005; Cardaropoli et al. 2012).
Decidual Endometrium Mimics The Tumor Environment
Significant similarities exist between the proliferative, migratory, and invasive properties of placental cells and cancer cells, with shared molecular circuits (Ferretti et al. 2007; Genbacev et al. 1997). The penetrative nature of hemochorial placentation mimics the phenomena observed with highly invasive tumors (Strickland and Richards 1992). A study on the tumor suppressor gene maspin revealed that it is differentially regulated in cytotrophoblasts during human placental development, with its downregulation being critical during implantation and early placental development (Dokras et al. 2002). The process of decidualization in the first trimester of pregnancy occurs in physiological hypoxia in the uterine endometrium, thus favoring leukocyte recruitment. A similarly hypoxic environment exists at sites of tumor development, and MФ recruitment (M2) occurs in tumor tissues in response to cytokine secretion by tumor cells. M2MФs further help tumor cell growth and angiogenesis by secreting VEGF. There may be increased production of VEGF by MФs in preeclamptic cases, which leads to vascular dysfunction and severe disease (Fan et al. 2014). Further study is needed to unravel the link between MФs and VEGF production in normal pregnancy and in pregnancy complications, as well as the involvement of macrophages in the pathogenesis of preeclampsia.
Mouse Models for Studying MФ Function in Pregnancy
MФs are involved in various functions depending on their anatomical locations and body physiology. The mechanisms behind their function are of great scientific interest; however, the unavailability of sufficient numbers of primary tissue MФs with high purity makes the study of this population difficult. Therefore, mouse models play vital roles in the study of MФ functions. Their functional role in reproduction has been revealed to some extent by studies in the osteopetrotic Csfop/Csfop mouse (having a naturally occurring Csf-null mutation). The CSF-1 (M-CSF) molecule is the major stimulant of MФ proliferation, differentiation, and recruitment. The Csfop/Csfop mice severely lack MФs in many tissues, with the ovary and uterus being almost devoid of them (Wiktor-Jedrzejczak et al. 1990). They show extended estrous cycles and lower ovulation rates due to impaired function of the hypothalamic–pituitary–gonadal axis and intra-ovarian defects (Cohen et al. 2002). The CSF-1 null mutants have drawbacks for the study of MФ functions in the establishment and maintenance of pregnancy because cytokines and chemokines other than M-CSF also play roles in recruiting MФs to the ovary and uterus and as a decreased proportion of these animals have normal pregnancies. The density of F4/80+ MФs in these mice is about 53% of the wild type level, and in corpora lutea, 35% of the wild type level on day 1 post-coitum (Pollard et al. 1998; Robertson et al. 1998). The transgenic mouse Cd11b-Dtr provides a reliable approach to the study of MФ function in pregnancy (Duffield et al. 2005). These mice show severe depletion of CD11b+ MФs in systemic circulation after administration of diphtheria toxin (DT) at nanogram levels. Care et al. (2013) have reported that acute depletion of CD11b+ MФs during early pregnancy in these mice results in a complete failure of implantation which is alleviated by administration of DT-resistant, wild type MФs. MФs also play a crucial role in regulating CL development during embryo implantation in mice (Care et al. 2013). Fejer et al. (2013) have developed a method yielding self-renewing, nontransformed, GM-CSF/STAT5—dependent MФs (Max Planck Institute cells) from the mouse fetal liver, reflecting innate immune status of alveolar MФs (Fejer et al. 2013).
Therapeutic Targeting of MФs in Pregnancy‑Related Disorders
MФs are currently getting attention as therapeutic targets. Various nanoparticles have been used to deliver therapeutic agents to MФs in the body to treat genetic disorders and persistent infections, induce MФ death, regulate accessory functions of MФs, and diagnose pathologic conditions, like cancer (Moghimi et al. 2005). Particulate systems, such as liposomes, polymeric micelles, and polymeric micro- and nanoparticles contribute efficiently to the delivery of therapeutic agents to MФs in different physiological portals of entry (Moghimi et al. 2012). MФ-derived foam cells have emerged as therapeutic targets in atherosclerosis, and folate receptor-mediated targeting of therapeutic and imaging agents to activated MФs might play a role in destroying FR (folate receptor)-expressing MФs in rheumatoid arthritis (Choudhury et al. 2005; Paulos et al. 2004). There is great interest in cancer biology for MФs to be used as therapeutic targets. The TAMs have been shown to modulate the effects of various anti-cancer therapies and promote tumor growth and angiogenesis. Preclinical studies have shown ways to polarize TAMs from the M2 to the M1 type in tumors so that the macrophages phagocytose tumor cells (De Palma and Lewis 2013). One of these strategies includes the use of histidine-rich glycoprotein (HRG), which induces downregulation of MФ PlGF, promotes blood vessel normalization, and increases the delivery and efficacy of chemotherapy in tumor models in mice (Rolny et al. 2011). Another strategy involves nuclear factor κB signaling, or exposure of TAMs to anti-IL10R antibodies combined with the TLR-9 ligand CpG (Hagemann et al. 2008; Guiducci et al. 2005). This strategy was shown to induce hemorrhagic tumor necrosis, activate DCs and cytotoxic T cells, and cause tumor remnant clearance. Trabectedin, a recent chemotherapeutic agent, has been observed to deplete monocytes/MФs in the blood, spleen, and tumors and to reduce angiogenesis in mouse models (Germano et al. 2013).
Some pregnancy complications are linked with M1/M2 imbalances in early inflammatory phase of pregnancy. It seems that the M1 subtype predominates over the M2 subtype in those cases, such as spontaneous abortions and disorders caused by inadequate spiral artery remodeling (Brown et al. 2014). Strategies to manipulate MФ polarity at the maternal–fetal interface and shift the MФ balance from M1 to M2 may alleviate pregnancy complications. In preeclampsia, the number of non-classical monocytes increases, which may promote inflammation (Faas et al. 2014). It has been observed that in preeclampsia, there are fewer M2MФs (Martinez et al. 2009). Instead, M1MФs predominate in preeclampsia, as severe inflammatory conditions prevail in maternal–fetal interface. The process of M1–M2MФ type polarization may be the therapeutic target for treatment of preeclampsia.
Conclusions and Future Directions
Decidual MФs play significant roles in successful pregnancy, with wide plasticity according to the environmental milieu. M1MФs (proinflammatory) promote embryo implantation in early pregnancy and are actively involved in protection of the fetus from intrauterine infection, cervical ripening, and parturition. M2MФs provide an immu-notolerant environment for the semiallogeneic fetus to be carried in the uterus throughout pregnancy. Treg cells also promote an immune-suppressive environment by suppressing Teffector cells through cell–cell interactions with other immune cells, like M2MФs and DCs. Many pregnancy complications, such as preeclampsia, IUGR, and RSA, have indications of abnormal MФ responses. Extensive studies have been done to unravel the cell response in different types of cancers and pregnancy complications using mouse models. The switching of MФ polarity could be a potential therapeutic target to treat pregnancy complications, as is predicted for cancer treatment. These cells have great potential to be targeted and exploited for treatment of pregnancy complications as well as for cancer therapy in the future. The signaling pathways (which induce the polarization) can also be targeted to achieve the required phenotype of MФ and recovery from the diseased condition. The plasticity property of MФs could be exploited for the development of therapeutics in pregnancy complications. Future investigations regarding decidual MФs and their roles in physiological and pathological conditions through system biology approaches will give direction to the development of therapeutics.
Acknowledgements
We thank Matt Petitt, PhD, for his comments and editorial assistance in preparation of the manuscript.
Funding The present work was supported by the NICHD award number R01HD088549 to Dr. Nihar R Nayak.
Footnotes
Conflict of interest The authors declare that they have no competing interests.
Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- Abrahams VM, Kim YM, Straszewski SL, Romero R, Mor G (2004) Macrophages and apoptotic cell clearance during pregnancy. Am J Reprod Immunol 51:275–282 [DOI] [PubMed] [Google Scholar]
- Amjadi F, Salehi E, Mehdizadeh M, Aflatoonian R (2014) Role of the innate immunity in female reproductive tract. Adv Biomed Res 3:1Arcuri F, Cintorino M, Vatti R, Carducci A, Liberatori S, Paulesu L (1999) Expression of macrophage migration inhibitory factor transcript and protein by first trimester human trophoblasts. Biol Reprod 60(6):1299e303. [DOI] [PubMed] [Google Scholar]
- Arcuri F, Buchwalder L, Toti P et al. (2007) Differential regulation of colony stimulating factor 1 and macrophage migration inhibitory factor expression by inflammatory cytokines in term human decidua: implications for macrophage trafficking at the fetal-maternal interface. Biol Reprod 76(3):433e9. [DOI] [PubMed] [Google Scholar]
- Aschkenazi S, Straszewski S, Verwer KM, Foellmer H, Rutherford T, Mor G (2002) Differential regulation and function of the Fas/Fas ligand system in human trophoblast cells. Biol Reprod 66:1853–1861Bale TL, Vale WW (2004) CRF and CRF receptors: role in stress responsivity and other behaviors. Annu Rev Pharmacol Toxicol 44:525–557 [Google Scholar]
- Bevilacqua E, Paulesu L, Ferro EA, Ietta F, Faria MR, Lorenzon AR, Costa AF, Martucci M (2014) Review: putative roles for the macrophage migratory inhibitory factor at the maternal fetal interface. Placenta 35(Suppl):S51–S56 [DOI] [PubMed] [Google Scholar]
- Blois SM, Alba Soto CD, Tometten M, Klapp BF, Margni RA, Arck PC (2004) Lineage, maturity, and phenotype of uterine murine dendritic cells throughout gestation indicate a protective role in maintaining pregnancy. Biol Reprod 70:1018–1023 [DOI] [PubMed] [Google Scholar]
- Bornstein P (1995) Diversity of function is inherent in matricellular proteins: an appraisal of thrombospondin 1. J Cell Biol 130:503–506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown MB, von Chamier M, Allam AB, L Reyes (2014) M1/M2 macrophage polarity in normal and complicated pregnancy. Front Immunol 5:606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bucala R (2007) MIF: the most interesting factor. World Scientific Publishing Co.; p, Singapore, p 332 [Google Scholar]
- Burk MR, Troeger C, Brinkhaus R, Holzgreve W, Hahn S (2001) Severely reduced presence of tissue macrophages in the basal plate of pre-eclamptic placentae. Placenta 22:309–316 [DOI] [PubMed] [Google Scholar]
- Burrows TD, King A, Loke YW (1996) Trophoblast migration during human placental implantation. Hum Reprod Update 2:307–321 [DOI] [PubMed] [Google Scholar]
- Calandra T, Roger T (2003) Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immunol 3(10):791e800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardaropoli S, Paulesu L, Romagnoli R et al. (2012) Macrophage migration inhibitory factor in fetoplacental tissues from preeclamptic pregnancies with or without fetal growth restriction. Clin Dev Immunol 2012:639342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Care AS, Diener KR, Jasper MJ, Brown HM, Ingman WV, Robertson SA (2013) Macrophages regulate corpus luteum development during embryo implantation in mice. J Clin Invest 123(8):3472–3487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carruba G, D’Agostino P, Miele M et al. (2003) Estrogen regulates cytokine production and apoptosis in PMA-differentiated, macrophage-like U937 cells. J Cell Biochem 90:187–196 [DOI] [PubMed] [Google Scholar]
- Chen G, Tan CS, The BK, Lu J (2011) Molecular mechanisms for synchronized transcription of three complement C1q subunit genes in dendritic cells and macrophages. J Biol Chem 286(40):34941–34950 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choudhury RP, Lee JM, Greaves DR (2005) Mechanisms of disease: macrophage-derived foam cells emerging as therapeutic targets in atherosclerosis. Nat Clin Pract Cardiovasc Med 2(6):309–315 [DOI] [PubMed] [Google Scholar]
- Clark DE, Smith SK, Licence D, Evans AL, Charnock-Jones DS (1998) Comparison of expression patterns for placenta growth factor, vascular endothelial growth factor (VEGF), VEGF-B and VEGF-C in the human placenta throughout gestation. J Endocrinol 159:459–467 [DOI] [PubMed] [Google Scholar]
- Clauss M, Weich H, Breier G, Knies U, Röckl W, Waltenberger J, Risau W (1996) The vascular endothelial growth factor receptor Flt-1 mediates biological activities. Implications for a functional role of placenta growth factor in monocyte activation and chemotaxis. J Biol Chem 271(30):17629–17634 [DOI] [PubMed] [Google Scholar]
- Co EC, Gormley M, Kapidzic M, Rosen DB, Scott MA, Stolp HA, McMaster M, Lanier LL, Bárcena A, Fisher SJ (2013) Maternal decidual macrophages inhibit NK cell killing of invasive cytotrophoblasts during human pregnancy. Biol Reprod 20 88(6):155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coffelt SB, Hughes R, Lewis CE (2009) Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta 1796:11–18 [DOI] [PubMed] [Google Scholar]
- Cohen PE, Zhu L, Nishimura K, Pollard JW (2002) Colony-stimulating factor 1 regulation of neuroendocrine pathways that control gonadal function in mice. Endocrinology 143(4):1413–1422 [DOI] [PubMed] [Google Scholar]
- Cupurdija K, Azzola D, Hainz U, Gratchev A, Heitger A, Takikawa O, Goerdt S, Wintersteiger R, Dohr G, Sedlmayr P (2004) Macrophages of human first trimester decidua express markers associated to alternative activation. Am J Reprod Immunol 51(2):117–122 [DOI] [PubMed] [Google Scholar]
- Dakic A, Metcalf D, DiRago L, Mifsud S, Wu L, Nutt SL (2005) PU.1 regulates the commitment of adult hematopoietic progenitors and restricts granulopoiesis. J Exp Med 201:1487–1502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- David Dong ZM, Aplin AC, Nicosia RF (2009) Regulation of angiogenesis by macrophages, dendritic cells, and circulating myelomonocytic cells. Curr Pharm Des 15:365–379 [DOI] [PubMed] [Google Scholar]
- De Palma M, Lewis CE (2013) Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 23(3):277–286 [DOI] [PubMed] [Google Scholar]
- Dechaud H, Maudelonde T, Daures JP, Rossi JF, Hedon B (1998) Evaluation of endometrial inflammation by quantification of macrophages, T lymphocytes, and interleukin-1 and −6 in human endometrium. J Assist Reprod Genet 15:612–618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeLoia JA, Stewart-Akers AM, Brekosky J, Kubik CJ (2002) Effects of exogenous estrogen on uterine leukocyte recruitment. Fertil Steril 77:548–554 [DOI] [PubMed] [Google Scholar]
- Dokras A, Gardner LM, Kirschmann DA, Seftor EA, Hendrix MJ (2002) The tumour suppressor gene maspin is differentially regulated in cytotrophoblasts during human placental development. Placenta 23(4):274–280 [DOI] [PubMed] [Google Scholar]
- Douglas NC, Zimmermann RC, Tan QK, Sullivan-Pyke CS, Sauer MV, Kitajewski JK, Shawber CJ (2014) VEGFR-1 blockade disrupts peri-implantation decidual angiogenesis and macrophage recruitment. Vasc Cell 6:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duffield JS et al. (2005) Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest 115(1):56–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duncan WC (2000) The human corpus luteum: remodelling during luteolysis and maternal recognition of pregnancy. Rev Reprod 5:12–17 [DOI] [PubMed] [Google Scholar]
- Faas MM, Spaans F, De Vos P (2014) Monocytes and macrophages in pregnancy and pre-eclampsia. Front Immunol 5:298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faas MM, de Vos P (2017) Uterine NK cells and macrophages in pregnancy. Placenta 56:44–52 [DOI] [PubMed] [Google Scholar]
- Fan X, Rai A, Kambham N, Sung JF, Singh N, Petitt M, Dhal S, Agrawal R, Sutton RE, Druzin ML, Gambhir SS, Ambati BK, Cross JC, Nayak NR (2014) Endometrial VEGF induces placental sFLT1 and leads to pregnancy complications. J Clin Invest 124(11):4941–4952 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fejer G, Wegner MD, Györy I (2013) Nontransformed, GM-CSF-dependent macrophage lines are a unique model to study tissue macrophage functions. Proc Natl Acad Sci USA 110(24):E2191–E2198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferretti C, Bruni L, Dangles-Marie V, Pecking AP, Bellet D (2007) Molecular circuits shared by placental and cancer cells, and their implications in the proliferative, invasive and migratory capacities of trophoblasts. Hum Reprod Update 13(2):121–141 [DOI] [PubMed] [Google Scholar]
- Fock V, Mairhofer M, Otti GR, Hiden U, Spittler A, Zeisler H, Fiala C, Knöfler M, Pollheimer J (2013) Macrophage-derived IL-33 is a critical factor for placental growth. J Immunol 1 191(7):3734–3743 [DOI] [PubMed] [Google Scholar]
- Galazka K, Wicherek L, Pitynski K, Kijowski J, Zajac K, Bednarek W, Dutsch-Wicherek M, Rytlewski K, Kalinka J, Basta A, Majka M (2009) Changes in the subpopulation of CD25 + CD4 + and FOXP3 + regulatory T cells in decidua with respect to the progression of labor at term and the lack of analogical changes in the subpopulation of suppressive B7-H4 macrophages–a preliminary report. Am J Reprod Immunol 61(2):136–146 [DOI] [PubMed] [Google Scholar]
- Gardner L, Moffett A (2003) Dendritic cells in the human decidua. Biol Reprod 69:1438–1446 [DOI] [PubMed] [Google Scholar]
- Genbacev O, Zhou Y, Ludlow JW, Fisher SJ (1997) Regulation of human placental development by oxygen tension. Science 277(5332):1669–1672 [DOI] [PubMed] [Google Scholar]
- Germano G, Frapolli R, Belgiovine C et al. (2013) Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 23(2):249–262 [DOI] [PubMed] [Google Scholar]
- Gnainsky Y, Granot I, Aldo PB, Barash A, Or Y, Schechtman E, Mor G, Dekel N (2010) Local injury of the endometrium induces an inflammatory response that promotes successful implantation. Fertil Steril 94(6):2030–2036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goetzl EJ, Banda MJ, Leppert D (1996) Matrix metalloproteinases in immunity. J Immunol 156:1–4 [PubMed] [Google Scholar]
- Gonzalez JM, Dong Z, Romero R, Girardi G (2011) Cervical remodeling/ripening at term and preterm delivery: the same mechanism initiated by different mediators and different effector cells. PLoS One 6(11):e26877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorczynski RM, Hadidi S, Yu G, Clark DA (2002) The same immunoregulatory molecules contribute to successful pregnancy and transplantation. Am J Reprod Immunol 48(1):18–26 [DOI] [PubMed] [Google Scholar]
- Gordon S (2003) Alternative activation of macrophages. Nat Rev Immunol 3:23–35 [DOI] [PubMed] [Google Scholar]
- Goto S, Ozaki Y, Suzumori N, Yasukochi A, Kawakubo T, Furuno T, Nakanishi M, Yamamoto K, Sugiura-Ogasawara M (2014) Role of cathepsin E in decidual macrophage of patients with recurrent miscarriage. Mol Hum Reprod 20(5):454–462 [DOI] [PubMed] [Google Scholar]
- Gough MJ, Melcher AA, Ahmed A et al. (2001) Macrophages orchestrate the immune response to tumor cell death. Cancer Res 61:7240–7247 [PubMed] [Google Scholar]
- Guiducci C, Vicari AP, Sangaletti S, Trinchieri G, Colombo MP (2005) Redirecting in vivo elicited tumor infiltrating macrophages and dendritic cells towards tumor rejection. Cancer Res 65:3437–3446 [DOI] [PubMed] [Google Scholar]
- Gustafsson C, Mjösberg J, Matussek A et al. (2008) Gene expression profiling of human decid-ual macrophages: evidence for immunosuppressive phenotype. PLoS One 3:e2078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hagemann T, Lawrence T, McNeish I, Charles KA, Kulbe H, Thomp-son RG, Robinson SC, Balkwill FR (2008) “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med 205:1261–1268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamilton S, Oomomian Y, Stephen G, Shynlova O, Tower CL, Garrod A, Lye SJ, Jones RL (2012) Macrophages infiltrate the human and rat decidua during term and preterm labor: evidence that decidual inflammation precedes labor. Biol Reprod 14 86(2):39. [DOI] [PubMed] [Google Scholar]
- Haney AF, Misukonis MA, Weinberg JB (1983) Macrophages and infertility: oviductal macrophages as potential mediators of infertility. Fertil Steril 39(3):310–315 [DOI] [PubMed] [Google Scholar]
- Heikkinen J, Mottonen M, Komi J, Alanen A, Lassila O (2003) Phenotypic characterization of human decidual macrophages. Clin Exp Immunol 131:498–505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Helige C, Ahammer H, Moser G, Hammer A, Dohr G, Huppertz B, Sedlmayr P (2014) Distribution of decidual natural killer cells and macrophages in the neighbourhood of the trophoblast invasion front: a quantitative evaluation. Hum Reprod 29(1):8–17 [DOI] [PubMed] [Google Scholar]
- Heloterä H, Alitalo K (2007) The VEGF family, the inside story. Cell 130(4):591–592 [DOI] [PubMed] [Google Scholar]
- Honig A, Rieger L, Kapp M, Sutterlin M, Dietl J, Kammerer U (2004) Indoleamine 2,3-dioxygenase (IDO) expression in invasive extravillous trophoblast supports role of the enzyme for materno-fetal tolerance. J Reprod Immunol 61:79–86 [DOI] [PubMed] [Google Scholar]
- Horcajadas JA, Pellicer A, Simón C (2007) Wide genomic analysis of human endometrial receptivity: new times, new opportunities. Hum Reprod Update 13:77–86 [DOI] [PubMed] [Google Scholar]
- Horton JS, Yamamoto SY, Bryant-Greenwood GD (2011) Relaxin modulates proinflammatory cytokine secretion from human decidual macrophages. Biol Reprod 85(4):788–797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houser BL (2012) Decidual macrophages and their roles at the maternal-fetal interface. Yale J Biol Med 85(1):105–118 [PMC free article] [PubMed] [Google Scholar]
- Houser BL, Tilburgs T, Hill J, Nicotra ML, Strominger JL (2011) Two unique human decidual macrophage populations. J Immunol 186(4):2633–2642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang SJ, Zenclussen AC, Chen CP (2010) The implication of aberrant GM-CSF expression in decidual cells in the pathogenesis of preeclampsia. Am J Pathol 177(5):2472–2482 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hunt JS (2006) Stranger in a strange land. Immunol Rev 213:36–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hunt JS, Petroff MG, Burnett TG (2000) Uterine leukocytes: key players in pregnancy. Semin Cell Dev Biol 11(2):127–137 [DOI] [PubMed] [Google Scholar]
- Huppertz B, Kingdom J, Caniggia I, Desoye G, Black S, Korr H, Kauf-mann P (2003) Hypoxia favours necrotic versus apoptotic shedding of placental syncytiotrophoblast into the maternal circulation. Placenta 24:181–190 [DOI] [PubMed] [Google Scholar]
- Huppertz B, Kadyrov M, Kingdom JC (2006) Apoptosis and its role in the trophoblast. Am J Obstet Gynecol 195(1):29–39 [DOI] [PubMed] [Google Scholar]
- Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA (2000) Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol 164:3596–3599 [DOI] [PubMed] [Google Scholar]
- Imhof BA, Aurrand-Lions M (2004) Adhesion mechanisms regulating the migration of monocytes. Nat Rev Immunol 4:432–444 [DOI] [PubMed] [Google Scholar]
- Iwasaki H, Somoza C, Shigematsu H et al. (2005) Distinctive and indispensable roles of PU.1 in maintenance of hematopoietic stem cells and their differentiation. Blood 106:1590–1600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaiswal MK, Mallers TM, Larsen B et al. (2012) V-ATPase up regulation during early pregnancy: a possible link to establishment of an inflammatory response during preimplantation period of pregnancy. Reproduction 143(5):713–725 [DOI] [PubMed] [Google Scholar]
- Jin Y, Wang X, Xiao Y, Lv C, Ding C, Lin Q (2009) The role of TSP-1 on decidual macrophages involved in the susceptibility to unexplained recurrent spontaneous abortion. Am J Reprod Immunol 61(3):253–260 [DOI] [PubMed] [Google Scholar]
- Johansson EL, Rudin A, Wassen L, Holmgren (1999) Distribution of lymphocytes and adhesion molecules in human cervix and vagina. Immunology 96:272–277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones RL, Hannan NJ, Kaitu’u TJ, Zhang J, Salamonsen LA (2004) Identification of chemokines important for leukocyte recruitment to the human endometrium at the times of embryo implantation and menstruation. J Clin Endocrinol Metab 89:6155–6167 [DOI] [PubMed] [Google Scholar]
- Karteris E, Vatish M, Hillhouse EW, Grammatopoulos DK (2005) Preeclampsia is associated with impaired regulation of the placental nitric oxide-cyclic guanosine monophosphate pathway by corticotropin-releasing hormone (CRH) and CRH-related peptides. J Clin Endocrinol Metab 90:3680–3687 [DOI] [PubMed] [Google Scholar]
- Kats R, Al-Akoum M, Guay S, Metz C, Akoum A (2005) Cycle dependent expression of macrophage migration inhibitory factor in the human endometrium. Hum Reprod 20:3518–3525 [DOI] [PubMed] [Google Scholar]
- Keenihan SN, Robertson SA (2004) Diversity in phenotype and steroid hormone dependence in dendritic cells and macrophages in the mouse uterus. Biol Reprod 70:1562–1572 [DOI] [PubMed] [Google Scholar]
- Keskin DB, Allan DS, Rybalov B et al. (2007) TGF beta promotes conversion of CD16+ peripheral blood NK cells into CD16- NK cells with similarities to decidual NK cells. Proc Natl Acad Sci USA 104:3378–3383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JS, Romero R, Cushenberry E, Kim YM, Erez O, Nien JK, Yoon BH, Espinoza J, Kim CJ (2007) Distribution of CD14 + and CD68 + macrophages in the placental bed and basal plate of women with preeclampsia and preterm labor. Placenta 28(5–6):571–576 [DOI] [PubMed] [Google Scholar]
- King A (2000) Uterine leukocytes and decidualization. Hum Reprod Update 6:28–36 [DOI] [PubMed] [Google Scholar]
- King A et al. (2000a) HLA-E is expressed on trophoblast and interacts with CD94/NKG2 receptors on decidual NK cells. Eur J Immunol 30:1623–1631 [DOI] [PubMed] [Google Scholar]
- King A et al. (2000b) Surface expression of HLA-C antigen by human extravillous trophoblast. Placenta 21:376–387 [DOI] [PubMed] [Google Scholar]
- Kitaya K, Yamaguchi T, Yasuo T, Okubo T, Honjo H (2007) Post-ovulatory rise of endometrial CD16(−) natural killer cells: in situ proliferation of residual cells or selective recruitment from circulating peripheral blood? J Reprod Immunol 76:45–53 [DOI] [PubMed] [Google Scholar]
- Kudo Y, Boyd CAR, Spyropoulou I, Redman CWG, Takikawa O, Katsuki T, Hare T, Ohama K, Sargent IL (2004) Indoleamine 2,3-dioxygenase: distribution and function in the developing human placenta. J Reprod Immunol 61:87–98 [DOI] [PubMed] [Google Scholar]
- Lafayette RA, Hladunewich MA, Derby G, Blouch K, Druzin ML, Myers BD (2011) Serum relaxin levels and kidney function in late pregnancy with or without preeclampsia. Clin Nephrol 75:226–232 [DOI] [PubMed] [Google Scholar]
- Laskarin G, Cupurdija K, Tokmadzic VS et al. (2005) The presence of functional mannose receptor on macrophages at the maternal-fetal interface. Hum Reprod 20(4):1057–1066 [DOI] [PubMed] [Google Scholar]
- Lea RG, Clark DA (1991) Macrophages and migratory cells in endometrium relevant to implantation. Baillieres Clin Obstet Gynaecol 5(1):25–59 [DOI] [PubMed] [Google Scholar]
- Lee S, Chen TT, Barber CL et al. (2007) Autocrine VEGF signaling is required for vascular homeostasis. Cell 130(4):591–592 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JY, Lee M, Lee SK (2011) Role of endometrial immune cells in implantation. Clin Exp Reprod Med 38(3):119–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee SK, Kim CJ, Kim DJ, Kang JH (2015) Immune cells in the female reproductive tract. Immune Netw 15(1):16–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leung PH, Salamonsen LA, Findlay JK (1998) Immunolocalization of inhibin and activin subunits in human endometrium across the menstrual cycle. Hum Reprod 13:3469–3477 [DOI] [PubMed] [Google Scholar]
- Li C, Houser BL, Nicotra ML, Strominger JL (2009) HLA-G homodimer-induced cytokine secretion through HLA-G receptors on human decidual macrophages and natural killer cells. Proc Natl Acad Sci USA 106:5767–5772 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li ZH, Wang LL, Liu H et al. (2019) Galectin-9 alleviates LPS-induced preeclampsia-like impairment in rats via switching decidual macrophage polarization to M2 subtype. Front Immunol 9:3142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lidström C, Matthiesen L, Berg G, Sharma S, Ernerudh J, Ekerfelt C (2003) Cytokine secretion patterns of NK cells and macrophages in early human pregnancy decidua and blood: implications for suppressor macrophages in decidua. Am J Reprod Immunol 50:444–452 [DOI] [PubMed] [Google Scholar]
- Lockwood CJ, Matta P, Krikun G, Koopman LA, Masch R, Toti P, Arcuri F, Huang ST, Funai EF, Schatz F (2006) Regulation of monocyte chemoattractant protein-1 expression by tumor necrosis factor-alpha and interleukin-1beta in first trimester human decidual cells: implications for preeclampsia. Am J Pathol 168(2):445–452 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lombardelli L, Aguerre-Girr M, Logiodice F, Kullolli O, Casart Y, Polgar B, Berrebi A, Romagnani S, Maggi E, Le Bouteiller P, Piccinni MP (2013) HLA-G5 induces IL-4 secretion critical for successful pregnancy through differential expression of ILT2 receptor on decidual CD4 + T cells and macrophages. J Immunol 191(7):3651–3662 [DOI] [PubMed] [Google Scholar]
- Madhukaran SP, Kishore U, Jamil K, Teo BH, Choolani M, Lu J (2015) Transcriptional factor PU.1 regulates decidual C1q expression in early pregnancy in human. Front Immunol 6:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Manaster I, Mandelboim O (2010) The unique properties of uterine NK cells. Am J Reprod Immunol 63:434–444 [DOI] [PubMed] [Google Scholar]
- Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M (2004) The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25:677–686 [DOI] [PubMed] [Google Scholar]
- Martinez FO, Sica A, Mantovani A, Locati M (2008) Macrophage activation and polarization. Front Biosci 13:453–461 [DOI] [PubMed] [Google Scholar]
- Martinez FO, Helming L, Gordon S (2009) Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 27:451–483 [DOI] [PubMed] [Google Scholar]
- McIntire RH, Ganacias KG, Hunt JS (2008) Programming of human monocytes by the uteroplacental environment. Reprod Sci 15(5):437–447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM (2000) M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol 164:6166–617310843666 [Google Scholar]
- Miwa N, Hayakawa S, Miyazaki S, Myojo S, Sasaki Y, Sakai M, Takikawa O, Saito S (2005) IDO expression on decidual and peripheral blood dendritic cells and monocytes/macrophages after treatment with CTLA-4 or interferon-gamma increase in normal pregnancy but decrease in spontaneous abortion. Mol Hum Reprod 11(12):865–870 [DOI] [PubMed] [Google Scholar]
- Mizuno M, Aoki K, Kimbara T (1994) Functions of macrophages in human decidual tissue in early pregnancy. Am J Reprod Immunol 31(4):180–188 [DOI] [PubMed] [Google Scholar]
- Moghimi SM, Hunter AC, Murray JC (2005) Nanomedicine: current progress and future prospects. FASEB J 19:311–330 [DOI] [PubMed] [Google Scholar]
- Moghimi SM, Parhamifar L, Ahmadvand D, Wibroe PP, Andresen TL, Farhangrazi ZS, Hunter AC (2012) Particulate systems for targeting of macrophages: basic and therapeutic concepts. J Innate Immun 4(5–6):509–528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mor G, Abrahams VM (2003) Potential role of macrophages as immu-noregulators of pregnancy. Reprod Biol Endocrinol 1:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mor G, Cardenas I, Abrahams V, Guller S (2011) Inflammation and pregnancy: the role of the immune system at the implantation site. Ann NY Acad Sci 1221:80–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mosser DM (2003) The many faces of macrophage activation. J Leukoc Biol 73:209–212 [DOI] [PubMed] [Google Scholar]
- Munn DH, Sharma MD, Lee JR et al. (2002) Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase. Science 297:1867–1870 [DOI] [PubMed] [Google Scholar]
- Nagamatsu T, Schust DJ (2010a) The contribution of macrophages to normal and pathological pregnancies. Am J Reprod Immunol 63:460–471 [DOI] [PubMed] [Google Scholar]
- Nagamatsu T, Schust DJ (2010b) The immunomodulatory roles of macrophages at the maternal-fetal interface. Reprod Sci 17:209–218 [DOI] [PubMed] [Google Scholar]
- Nakamura H, Jasper MJ, Hull ML, Aplin JD, Robertson SA (2012) Macrophages regulate expression of α 1, 2-fucosyltransferase genes in human endometrial epithelial cells. Mol Hum Reprod 18(4):204–215 [DOI] [PubMed] [Google Scholar]
- Neale D, Demasio K, Illuzi J, Chaiworapongsa T, Romero R, Mor G (2003) Maternal serum of women with pre-eclampsia reduces trophoblast cell viability: evidence for an increased sensitivity to Fas-mediated apoptosis. J Matern Fetal Neonatal Med 13:39–44 [DOI] [PubMed] [Google Scholar]
- Ning F, Liu H, Lash GE (2016) The role of decidual macrophages during normal and pathological pregnancy. am j reprod Immunol 75(3):298–309 [DOI] [PubMed] [Google Scholar]
- Nishioku T, Hashimoto K, Yamashita K et al. (2002) Involvement of cathepsin E in exogenous antigen processing in primary cultured murine microglia. J Biol Chem 277:4816–4822 [DOI] [PubMed] [Google Scholar]
- Palani S, Maksimow M, Miiluniemi M, Auvinen K, Jalkanen S, Salmi M (2011) Stabilin-1/CLEVER-1, a type 2 macrophage marker, is an adhesion and scavenging molecule on human placentalmacrophages. Eur J Immunol 41(7):2052–2063 [DOI] [PubMed] [Google Scholar]
- Parry RV, Chemnitz JM, Frauwirth KA et al. (2005) CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 25:9543–9553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pate JL, Landis Keyes P (2001) Immune cells in the corpus luteum: friends or foes? Reproduction 122:665–676 [DOI] [PubMed] [Google Scholar]
- Paulos CM, Turk MJ, Breur GJ, Low PS (2004) Folate receptor-mediated targeting of therapeutic and imaging agents to activated macrophages in rheumatoidarthritis. Adv Drug Deliv Rev 56(8):1205–1217 [DOI] [PubMed] [Google Scholar]
- Pearce BD, Garvin SE, Grove J et al. (2008) Serum macrophage migration inhibitory factor in the prediction of preterm delivery. Am J Obstet Gynecol 199(1):46.e1e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pearce BD, Grove J, Bonney EA et al. (2010) Interrelationship of cytokines, hypothalamic-pituitary-adrenal axis hormones, and psychosocial variables in the prediction of preterm birth. Gynecol Obstet Invest 70(1):40e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Penny LA (2000) Monocyte chemoattractant protein 1 in luteolysis. Rev Reprod 5:63–66 [DOI] [PubMed] [Google Scholar]
- Petroff MG, Sedlmayr P, Azzola D, Hunt JS (2002) Decidual macrophages are potentially susceptible to inhibition by class Ia and class Ib HLA molecules. J Reprod Immunol 56(1–2):3–17 [DOI] [PubMed] [Google Scholar]
- Petrova TV, Makinen T, Alitalo K (1999) Signaling via vascular endothelial growth factor receptors. Exp Cell Res 253(1):117–130 [DOI] [PubMed] [Google Scholar]
- Petsas G, Jeschke U, Richter DU, Minas V, Hammer A, Kalantari-dou S, Toth B, Tsatsanis C, Friese K, Makrigiannakis A (2012) Aberrant expression of corticotropin-releasing hormone in preeclampsia induces expression of FasL in maternal macrophages and extravillous trophoblast apoptosis. Mol Hum Reprod 18(11):535–545 [DOI] [PubMed] [Google Scholar]
- Plaks V, Birnberg T, Berkutzki T et al. (2008) Uterine DCs are crucial for decidua formation during embryo implantation in mice. J Clin Invest 118:3954–3965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollard JW (1997) Role of colony-stimulating factor-1 in reproduction and development. Mol Reprod Dev 46:54–60 [DOI] [PubMed] [Google Scholar]
- Pollard JW (1998) Role of cytokines in the pregnant uterus of interstitial implanting species In: Bazer FW (ed) The endocrinology of pregnancy. Humana Press Inc., Totowa, pp 59–82 [Google Scholar]
- Pollard JW (2008) Uterine DCs are essential for pregnancy. J Clin Invest 118:3832–3835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollard JW, Lin EY, Zhu L (1998) Complexity in uterine macrophage responses to cytokines in mice. Biol Reprod 58(6):1469–1475 [DOI] [PubMed] [Google Scholar]
- Porcheray F, Viaud S, Rimaniol AC, Léone C, Samah B, Dereuddre-Bosquet N, Dormont D, Gras G (2005) Macrophage activation switching: an asset for the resolution of inflammation. Clin Exp Immunol 142(3):481–489 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiu X, Zhu L, Pollard JW (2009) Colony-Stimulating factor-1-dependent macrophage functions regulate the maternal decidua immune responses against Listeria monocytogenes infections during early gestation in mice. Infect Immun 77(1):85–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Quillay H, El Costa H, Marlin R et al. (2015) Distinct characteristics of endometrial and decidual macrophages and regulation of their permissivity to HIV-1 infection by SAMHD1. J Virol 89(2):1329–1339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rami D, La Bianca M, Agostinis C, Zauli G, Radillo O, Bulla R (2014) The first trimester gravid serum regulates procalcitonin expression in human macrophages skewing their phenotype in vitro. Mediat Inflamm 2014:1–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reister F, Frank HG, Heyl W et al. (1999) The distribution of macrophages in spiral arteries of the placental bed in pre-eclampsia differs from that in healthy patients. Placenta 20:229–233 [DOI] [PubMed] [Google Scholar]
- Reister F, Frank HG, Kingdom JC et al. (2001) Macrophage-induced apoptosis limits endovascular trophoblast invasion in the uterine wall of preeclamptic women. Lab Invest 81:1143–1152 [DOI] [PubMed] [Google Scholar]
- Renaud SJ, Graham CH (2008) The role of macrophages in uteroplacental interactions during normal and pathological pregnancy. Immunol Invest 37:535–564 [DOI] [PubMed] [Google Scholar]
- Robertson SA, Allanson M, Mau VJ (1998) Molecular regulation of uterine leukocyte recruitment during early pregnancy in the mouse. Placenta 19(suppl 1):101–119 [Google Scholar]
- Rolny C, Mazzone M, Tugues S et al. (2011) HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through down regulation of PlGF. Cancer Cell 19:31–44 [DOI] [PubMed] [Google Scholar]
- Saito S, Sakai M (2003) Th1/Th2 balance in preeclampsia. J Reprod Immunol 59:161–173 [DOI] [PubMed] [Google Scholar]
- Salamonsen LA, Lathbury LJ (2000) Endometrial leukocytes and menstruation. Hum Reprod Update 6:16–27 [DOI] [PubMed] [Google Scholar]
- Sayama S, Nagamatsu T, Schust DJ, Itaoka N, Ichikawa M, Kawana K, Yamashita T, Kozuma S, Fujii T (2013) Human decidual macrophages suppress IFN-γ production by T cells through costimulatory B7-H1:PD-1 signaling in early pregnancy. J Reprod Immunol 100(2):109–117 [DOI] [PubMed] [Google Scholar]
- Schonkeren D, van der Hoorn ML, Khedoe P, Swings G, van Beelen E, Claas F, van Kooten C, de Heer E, Scherjon S (2011) Differential distribution and phenotype of decidual macrophages in preeclamptic versus control pregnancies. Am J Pathol 178(2):709–717 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schrocksnadel H, Baier-Bitterlich G, Dapunt O, Wacher H, Fuchs D (1996) Decreased plasma tryptophan in pregnancy. Obstet Gynecol 88:47–50 [DOI] [PubMed] [Google Scholar]
- Sedlmayr P, Blaschitz A, Wintersteiger R, Semlitsch M, Hammer A, MacKenzie CR, Walcher W, Reich O, Takikawa O, Dohr G (2002) Localization of indoleamine 2,3-dioxygenase in human female reproductive organs and placenta. Mol Hum Reprod 8:385–391 [DOI] [PubMed] [Google Scholar]
- Selvaraj SK, Giri RK, Perelman N, Johnson C, Malik P, Kalra VK (2003) Mechanism of monocyte activation and expression of proinflammatory cytochemokines by placenta growth factor. Blood 102(4):1515–1524 [DOI] [PubMed] [Google Scholar]
- Shakhawat A, Shaikly V, Elzatma E, Mavrakos E, Jabeen A, Fernán-dez N (2010) Interaction between HLA-G and monocyte/macrophages in human pregnancy. J Reprod Immunol 85(1):40–46 [DOI] [PubMed] [Google Scholar]
- Shin M, Kadowaki T, Iwata J, Kawakubo T, Yamaguchi N, Takii R, Tsukuba T, Yamamoto K (2007) Association of cathepsin E with tumor growth arrest through angiogenesis inhibition and enhanced immune responses. Biol Chem 388:1173–1181 [DOI] [PubMed] [Google Scholar]
- Shobu T et al. (2006) The surface expression of HLA-F on decidual trophoblasts increases from mid to term gestation. J Reprod Immunol 72:18–32 [DOI] [PubMed] [Google Scholar]
- Singh U, Nicholson G, Urban BC, Sargent IL, Kishore U, Bernal AL (2005) Immunological properties of human decidual macrophages—a possible role in intrauterine immunity. Reproduction 129:631–637 [DOI] [PubMed] [Google Scholar]
- Smith SD, Dunk CE, Aplin JD, Harris LK, Jones RL (2009) Evidence for immune cell involvement in decidual spiral arteriole remodeling in early human pregnancy. Am J Pathol 174(5):1959–1971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stout RD, Suttles J (2004) Functional plasticity of macrophages: reversible adaptation to changing microenvironments. J Leukoc Biol 76:509–513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strickland S, Richards WG (1992) Invasion of the trophoblasts. Cell 71(3):355–357 [DOI] [PubMed] [Google Scholar]
- Sugino N, Kashida S, Karube-Harada A, Takiguchi S, Kato H (2002) Expression of vascular endothelial growth factor (VEGF) and its receptors in human endometrium throughout the menstrual cycle and in early pregnancy. Reproduction 123:379–387 [DOI] [PubMed] [Google Scholar]
- Svensson J, Jenmalm MC, Matussek A, Geffers R, Berg G, Ernerudh J (2011) Macrophages at the fetal-maternal interface express markers of alternative activation and are induced by M-CSF and IL-10. The J Immunol 187:3671–3682 [DOI] [PubMed] [Google Scholar]
- Svensson-Arvelund J, Ernerudh J (2015) The role of macrophages in promoting and maintaining homeostasis at the fetal-maternal interface. Am J Reprod Immunol 74(2):100–109 [DOI] [PubMed] [Google Scholar]
- Thiruchelvam U, Dransfield I, Saunders PT, Critchley HO (2013) The importance of the macrophage within the human endometrium. J Leukoc Biol 93:217–225 [DOI] [PubMed] [Google Scholar]
- Todros T, Bontempo S, Piccoli E et al. (2005) Increased levels of macrophage migration inhibitory factor (MIF) in preeclampsia. Eur J Obstet Gynecol Reprod Biol 123(2):162e6. [DOI] [PubMed] [Google Scholar]
- Tripathi C, Tewari BN, Kanchan RK, Baghel KS, Nautiyal N, Shrivastava R, Kaur H, Bhatt ML, Bhadauria S (2014) Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget 5(14):5350–5368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van den Heuvel M, Peralta C, Bashar S, Taylor S, Horrocks J, Croy BA (2005) Trafficking of peripheral blood CD56(bright) cells to the decidualizing uterus–new tricks for old dogmas? J Reprod Immunol 67:21–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Mourik MS, Heijnen CJ, Macklon NS (2009) Embryonic implantation: cytokines, adhesion molecules, and immune cells in establishing an implantation environment. J Leukoc Biol 85:4–19 [DOI] [PubMed] [Google Scholar]
- Verma S, Hiby SE, Loke YW, King A (2000) Human decidual natural killer cells express the receptor for and respond to the cytokine interleukin 15. Biol Reprod 62:959–968 [DOI] [PubMed] [Google Scholar]
- Verreck FA, de Boer T, Langenberg DM et al. (2004) Human IL-23-producing type 1 macrophages promote but IL-10- producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci USA 101:4560–4565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vogel P (2005) The current molecular phylogeny of Eutherian mammals challenges previous interpretations of placental evolution. Placenta 26(8–9):591–596 [DOI] [PubMed] [Google Scholar]
- von Rango U (2008) Fetal tolerance in human pregnancy— a crucial balance between acceptance and limitation of trophoblast invasion. Immunol Lett 115(1):21–32 [DOI] [PubMed] [Google Scholar]
- Wang WJ, Hao CF, Lin QD (2011) Dysregulation of macrophage activation by decidual regulatory T cells in unexplained recurrent miscarriage patients. J Reprod Immunol 92(1–2):97–102 [DOI] [PubMed] [Google Scholar]
- Wheeler KC, Jena MK, Pradhan BS, Nayak N, Das S, Hsu CD, Wheeler DS, Chen K, Nayak NR (2017) VEGF may contribute to macrophage recruitment and M2 polarization in the decidua. PLoS One 13(1):e0191040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiktor-Jedrzejczak W et al. (1990) Total absence of colony-stimulating factor 1 in the macrophage-deficient osteopetrotic (op/op) mouse. Proc Natl Acad Sci USA 87(12):4828–4832 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wildman DE, Chen C, Erez O, Grossman LI, Goodman M, Romero R (2006) Evolution of the mammalian placenta revealed by phylogenetic analysis. Proc Natl Acad Sci USA 103(9):3203–3208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams PJ, Bulmer JN, Searle RF, Innes BA, Robson SC (2009) Altered decidual leucocyte populations in the placental bed in pre-eclampsia and foetal growth restriction: a comparison with late normal pregnancy. Reproduction 138(1):177–184 [DOI] [PubMed] [Google Scholar]
- Wira CR, Fahey JV, Sentman CL, Pioli PA, Shen L (2005) Innate and adaptive immunity in female genital tract: cellular responses and interactions. Immunol Rev 206:306–335 [DOI] [PubMed] [Google Scholar]
- Wu ZM, Yang H, Li M, Yeh CC, Schatz F, Lockwood CJ, Di W, Huang SJ (2012) Pro-inflammatory cytokine-stimulated first trimester decidual cells enhance macrophage-induced apoptosis of extravillous trophoblasts. Placenta 33(3):188–194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xie X, He H, Colonna M, Seya T, Takai T, Croy BA (2005) Pathways participating in activation of mouse uterine natural killer cells during pregnancy. Biol Reprod 73:510–518 [DOI] [PubMed] [Google Scholar]
- Xu W, Roos A, Schlagwein N, Woltman AM, Daha MR, van Kooten C (2006) IL-10-producing macrophages preferentially clear early apoptotic cells. Blood 107:4930–4937 [DOI] [PubMed] [Google Scholar]
- Yamada H, Kato EH, Morikawa M et al. (2003) Decreased serum levels of macrophage migration inhibition factor in miscarriages with normal chromosome karyotype. Hum Reprod 18(3):616e20. [DOI] [PubMed] [Google Scholar]
- Yamauchi Y, Kuroki M, Imakiire T, Uno K, Abe H, Beppu R, Yamashita Y, Kuroki M, Shirakusa T (2002) Opposite effects of thrombospondin-1 via CD36 and CD47 on homotypic aggregation of monocytic cells. Matrix Biol 21:441–448 [DOI] [PubMed] [Google Scholar]
- Yang Z, Kong B, Mosser DM, Zhang X (2011) TLRs, macrophages, and NK cells: our understandings of their functions in uterus and ovary. Int Immunopharmacol 11(10):1442–1450 [DOI] [PubMed] [Google Scholar]
- Zhang X, Mosser DM (2008) Macrophage activation by endogenous danger signals. J Pathol 214:161–178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang YH, He M, Wang Y, Liao AH (2017) Modulators of the balance between M1 and M2 macrophages during Pregnancy. Front Immunol 9(8):120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Ma L, Hu X, Ji J, Mor G, Liao A (2019) The role of the PD-1/PD-L1 axis in macrophage differentiation and function during pregnancy. Hum Reprod 34(1):25–36 [DOI] [PubMed] [Google Scholar]
- Zhou Y, McMaster M, Woo K, Janatpour M, Perry J, Karpanen T, Alitalo K, Damsky C, Fisher SJ (2002) Vascular endothelial growth factor ligands and receptors that regulate human cytotrophoblast survival are dysregulated in severe preeclampsia and hemolysis, elevated liver enzymes, and low platelets syndrome. Am J Pathol 160:1405–1423 [DOI] [PMC free article] [PubMed] [Google Scholar]
