Pregnancy success requires constant dialogue between the mother and developing conceptus. Such crosstalk is facilitated through complex interactions between maternal and fetal cells at distinct tissue sites, collectively termed the “maternal–fetal interface.” The emergence of single-cell technologies has enabled a deeper understanding of the unique processes taking place at the maternal–fetal interface as well as the discovery of novel pathways and immune and nonimmune cell types. Single-cell approaches have also been applied to decipher the cellular dynamics throughout pregnancy, in parturition, and in obstetrical syndromes such as recurrent spontaneous abortion, preeclampsia, and preterm labor. Furthermore, single-cell technologies have been used during the recent COVID-19 pandemic to evaluate placental viral cell entry and the impact of SARS-CoV-2 infection on maternal and fetal immunity. In this brief review, we summarize the current knowledge of cellular immunobiology in pregnancy and its complications that has been generated through single-cell investigations of the maternal–fetal interface.

Pregnancy is a dynamic period of growth and development that is initiated at fecundation and culminates in parturition, resulting in the delivery of the conceptus (1). To successfully complete this journey, a coordinated series of complex events is required, which starts with the establishment of dialogue between the mother and embryo (29). Such communication is facilitated through multiple sites of maternal–fetal interaction, which have been collectively termed the “maternal–fetal interface” (1015). Such sites represent anatomically distinct areas of immunological contact, namely the decidua basalis, where maternal immune cells and decidual stromal cells (DSCs) interact with fetal extravillous trophoblasts (EVTs); the placental intervillous space, in which circulating maternal immune cells interact with the fetal syncytiotrophoblast (SCT); and the boundary where the decidua parietalis meets the chorion laeve in the chorioamniotic membranes (11, 13, 14, 16). Therefore, the maternal–fetal interface comprises numerous complex immune and nonimmune cellular interactions that support implantation (1721), promote fetal and placental development (22, 23), maintain homeostasis (17, 19, 2437), and participate in the inflammatory milieu implicated in parturition (3847). Accordingly, the disruption of maternal–fetal homeostasis at different stages of gestation has been implicated in multiple pathologies, ranging from pregnancy loss to preterm delivery (26, 27, 3032, 36, 37, 44, 45, 4851). Hence, deciphering the mechanisms of maternal–fetal crosstalk that lead to a normal term delivery as well as those implicated in obstetrical disease is paramount.

Recent investigations have leveraged the use of single-cell technologies to evaluate the transcriptome, proteome, and epigenome of the maternal–fetal interface at single-cell resolution. Among them, single-cell RNA sequencing (scRNA-seq) was the first to be described (5260) and has seen the most widespread use across reproductive science (6165). Indeed, scRNA-seq was used to provide the first insights into the cellular heterogeneity and interaction networks of the maternal–fetal interface in early (6677) and late (7880) gestation, and the resulting datasets of these pioneer studies have since been integrated into a number of subsequent investigations. Furthermore, single-cell technologies have been highly effective at uncovering the unique biological processes taking place at the maternal-fetal unit during midgestation (66, 8185) as well as those inflammatory pathways implicated in the processes of term and preterm parturition (80, 8690). Pertinent to the current pandemic, scRNA-seq has also provided important knowledge about the mechanisms of cell entry for SARS-CoV-2 as well as its deleterious effects on the maternal-fetal unit (9199). Herein, we aim to provide an overview of such studies in a succinct and comprehensive manner to foster future research that will utilize single-cell technologies to decipher the cellular mechanisms of disease in pregnancy complications.

The first trimester represents an important developmental window for the fetus, which relies heavily on proper placentation and spiral artery remodeling (100104). To aid in these processes, a carefully orchestrated series of interactions is required that involves maternal proangiogenic or tolerogenic immune cells; mesenchymal cells, endothelial cells, and DSCs; and the invading fetal trophoblasts. The first trimester has therefore been an attractive target period for investigating the cellular states, functions, and interactions that drive pregnancy establishment (6677). Moreover, because of the importance of a healthy first trimester for continued pregnancy, multiple single-cell studies have targeted the maternal–fetal interface during this stage to help unravel the mechanisms underlying pathologies such as recurrent spontaneous abortion (RSA) (71, 7375, 77) (Fig. 1). Below, we summarize the insights into the maternal-fetal immunobiology of the first trimester obtained by using single-cell technologies.

Placental cells in the first trimester

Some of the first studies of the early placenta used scRNA-seq to characterize the major cell types and their subsets present in this organ (6668). One survey used two scRNA-seq platforms (commercial 10x Genomics platform and a custom Drop-seq platform) to characterize the placental villi and found good correlation between these technologies (67). Two other investigations of the first-trimester maternal–fetal interface used either Smart-seq2 alone (66) or an integrated dataset obtained by using Smart-seq2 and the 10x Genomics platform (68). Placental cell types were largely consistent between studies, with villous cytotrophoblasts (VCTs), SCTs, EVTs, fibroblasts, endothelial cells, erythroblasts, and Hofbauer cells all being identified by their expression profiles (6669). Comparison of relative abundance indicated that, after trophoblast subsets, the most represented placental cell type was fibroblasts, followed by endothelial cells, erythroblasts, and Hofbauer cells (67), highlighting the capacity of scRNA-seq to map the cellular heterogeneity of the placenta.

In addition to their identification, the further characterization of trophoblast cell types revealed that cytotrophoblasts (CTBs) and EVTs include multiple subsets that displayed differing fusion potential or proliferative capacity, respectively (66). One of these CTB subsets displayed comparatively higher syncytin-2 expression (66), suggesting fusion competence that would allow SCT formation (105). Pseudotime analysis indicated a transition from CTB to EVT subtypes (66), consistent with other reports that proposed two trophoblast differentiation pathways by which VCT could transition to the EVT or SCT subsets (68). Such endpoint EVTs represented the most invasive trophoblast subset to be found in the maternal decidua (68) and characterized by expression of specific factors such as SOX4 (70). A recent investigation used scRNA-seq of the first-trimester placenta together with a prior dataset (68) to assess the trophoblast cell states implicated in the establishment, renewal, and differentiation of these cells (76). Four CTB states (CTB1–4), an SCT precursor-like CTB state, two column trophoblast (EVT precursor [106]) states, and an EVT state were identified on the basis of gene expression (76). Although the relative frequencies of the four CTB states increased throughout the first trimester, the column trophoblast and EVT states decreased from early to late first trimester, and SCT progenitors remained constant (76), highlighting the fluctuation of trophoblast cell states during this period. Combined RNA velocity and single-cell trajectory analyses were used to map primitive trophoblasts to the CTB2/CTB3 clusters, from which two putative differentiation pathways emerged: one directed toward EVT and the other toward an SCT precursor-like CTB state (76). To validate this finding, Monocle 3 and Slingshot were applied, and agreement between these two pseudotime analyses was demonstrated, indicating CTB2 as the closest point of origin (76). Thus, scRNA-seq allows the dissection of the origins and differentiation trajectories of the heterogeneous cell types that form the placenta.

It is worth mentioning the tools that have been developed for trajectory inference by which single cells are ordered on the basis of similarities in gene expression (107), as performed in the studies described above. A large number of trajectory inference methods have been developed, with more being generated often; yet, the largest difference among them is whether the topology is fixed and, if not, what kind of topology can be detected (107). A thorough comparison of current trajectory inference techniques indicated high variability between methods, thus it is clear that investigators should test multiple methods with their data, particularly when a priori knowledge of topology is lacking (107). Importantly, the effort required to identify and use such tools has been greatly diminished by the availability of resources and databases that can guide users to the best trajectory inference approaches (107109).

The characterization of other major placental cell types revealed that, in addition to trophoblasts, the fibroblast population includes multiple subsets, with two of these displaying expression profiles reminiscent of myofibroblasts (67), which could support the contractile properties of anchoring villi (110). Hofbauer cells were also found to include two subsets, one of which was found to express an activated, MHC class II (MHC-II)-expressing state that could be important for debris clearance in the developing placenta (66). Other cell types identified in the placenta include two distinct subsets of mesenchymal stromal cells, which displayed gene expression profiles indicative of participation in cell adhesion/migration or angiogenic processes, respectively (66).

The above-mentioned reports show that scRNA-seq represents a useful technology for uncovering the composition, differentiation pathways, and expression profiles of cell types in the placenta, a unique organ that carries out critical functions for fetal growth and development (101, 111). Importantly, such knowledge can be leveraged to unravel the molecular underpinnings of placental diseases.

Decidual cells in the first trimester

The first-trimester decidua represents a more heterogeneous cellular compartment than the placenta, given the increased abundance of immune cell types in addition to tissue cells (67, 68, 112). NK cells, APCs such as dendritic cells (DCs), macrophages, innate lymphoid cells, and T cells represent major subsets in the decidua, in addition to DSCs, fibroblasts, smooth muscle cells, endometrial epithelial cells, lymphatic endothelial cells, and vascular endothelial cells (67, 68, 112). The DSC compartment comprises multiple subsets characterized by their spatial location as well as their expression profiles (68, 71). Pseudotime analysis of DSCs and fibroblasts indicated that the fibroblast 1 subset can differentiate to either DSCs or fibroblast 2 (67). The differentiation of DSCs was further explored in a recent study of the murine uterus, in which the single-cell profiles of this tissue were compared between implantation sites (decidualized uterus) and interimplantation sites (nondecidualized uterus) (72). In this study, the decidual cellular compartment was subdivided into deep stromal, proliferating deep stromal, superficial stromal, intermediate decidual, and decidual cells, each with expression profiles indicative of distinct functionality (72). Two potential pseudotime differentiation pathways were determined: The first was a deep stromal to superficial stromal transition, and the second was a deep stromal to proliferative deep stromal to intermediate decidual/decidual transition, which was considered as corresponding to decidualization (72). Consistent with prior receptor–ligand interaction analyses in the decidua (67, 68), interaction networks between decidual cells and different major subsets such as immune cells, endothelial cells, and trophoblasts were also demonstrated (72). Interactions between decidual and immune cells included immunological pathways such as cytokine–cytokine receptor interaction, phagosome, NK cell–mediated cytotoxicity, and the NOD-like receptor signaling pathway, among others (72). Interestingly, interactions between decidual and endothelial cells showed substantial overlap with those between decidual and immune cells, as did the interactions between decidual cells and trophoblasts; notable terms shared among all three interactions included the TGF-β signaling pathway and the MAPK signaling pathway (72). Thus, decidualization involves a core set of signaling pathways that are shared among multiple cell types in this compartment, suggesting that this is a cooperative process. Expanding on this approach, a separate investigation used scRNA-seq data of the murine day 7.5 uterus to deconvolute spatial transcriptomic data derived from the same tissue, which allowed the assignment of single cells to specific uterine microenvironments (113). The integration of these datasets revealed the distinct distribution of different stromal subsets within the uterus as well as their clustering in relation to other cell types (113). Thus, the spatial localization of cell subsets at the maternal–fetal interface may provide insight into their functionality and potential interactions with neighboring cells.

The inference of cell–cell communications from scRNA-seq data has been an area of ongoing research. The first iteration of tools developed for this purpose was based on the expression of single receptor–ligand pairs (114116), which did not account for receptors that function as multiunit complexes. To overcome such a limitation, the CellPhoneDB version 2.0 tool was developed, which could predict enriched cell–cell signaling on the basis of minimum average expression of all members of multiunit complexes (117). More recently, this concept was further built upon by considering signaling cofactors such as soluble agonists/antagonists and membrane-bound coreceptors, which resulted in the creation of the cell–cell communication analysis tool CellChat (118). Although such databases provide a powerful new means of exploring scRNA-seq datasets, it should be considered that the resulting inferences are based solely on gene expression and thus require careful interpretation.

Other key players during early human pregnancy are decidual NK cells, which can be classified into different subsets based on single-cell clustering (67, 68, 73) or imaging mass cytometry (IMC) (112). Among these, a study reported that one NK subset (decidual NK cells, termed “dNK1”) is the most responsive to HLA-C/HLA-G and expresses more cytoplasmic granules than other subsets (68); thus, these decidual immune cells may be central for modulating EVT invasion during the first trimester. Pseudotime analysis of decidual NK cells indicated a potential differentiation pathway in which the endpoint subsets showed enrichment of similar immune-related processes (73). However, alternative pathways in which endpoint NK cells can return to an intermediate state have also been described (74). Decidual NK cells represent an important cellular contributor to pregnancy maintenance, given that a disruption in their functions (such as by poor maternal-fetal killer Ig-like receptor–HLA matching) has been linked to complications such as spontaneous abortion or preeclampsia (119122). A recent IMC-based investigation of the maternal–fetal interface identified six distinct clusters of NK cells (112). Consistent with a prior demonstration (68), a subset of CD69-expressing dNK1 cells was the most prominent in the first trimester (112). In addition to NK cells, a subset of T cells with a cytotoxic expression profile is also found in the first-trimester decidua (67), which may participate in the process of EVT invasion and remodeling. Other notable immune cell subsets at the early maternal–fetal interface include macrophages, reported to display single-cell gene expression profiles that are distinct from the conventional M1/M2 paradigm (75). The latter observation is consistent with the recently proposed role of decidual macrophages as homeostatic sentinels that promote fetal growth and sustain pregnancy in early (32) and late (37) gestation. Indeed, analysis of cell clustering based on IMC revealed that myeloid cells are most prominently localized to the trophoblast microenvironment as well as the microenvironment of other immune cells at the maternal–fetal interface (112). Moreover, among the six identified myeloid clusters, two were specifically enriched in the first trimester and lacked HLA-DR expression (112). These observations lend further support to the role of decidual macrophages as homeostatic mediators, even in the first trimester.

Taken together, these data provide deep insight into the cellular processes that characterize the first-trimester decidua. In particular, single-cell transcriptomic analysis is useful for uncovering the interactions between critical immune cells (such as NK cells) and stromal cells, which can inform the overall cellular dynamics of the maternal–fetal interface.

Cellular interactions at the maternal–fetal interface during the first trimester

A key strength of scRNA-seq is the capacity to infer cellular interactions within the target tissue by using analytical methods such as CellChat (118). Mapping of the mostly highly expressed receptor–ligand pairs among placental and decidual cells revealed prevalent cell–cell interactions taking place in these compartments (67, 68). Notable cell–cell interactions at the maternal–fetal interface include those between EVTs and maternal immune cells, which comprise signaling related to immunomodulation, growth, angiogenesis, and cell adhesion/recruitment (68). Yet, complex interaction networks also occur among the DSCs, fibroblasts, endothelial cells, NK cells, and macrophages to support cellular differentiation and homeostasis (67, 68). Moreover, cell–cell interaction analyses are not confined to a single tissue since studies incorporating both the decidua and placental villi have provided an overview of the strongest intercellular interactions between these compartments (67). A novel study mined bulk RNA-seq data from the first-trimester decidua and placental villi to identify sexually dimorphic receptor–ligand pairs that could influence cellular interactions in these compartments (69). Subsequent scRNA-seq identified the top sexually dimorphic genes within major cell-type clusters (e.g., trophoblasts, stromal cells, Hofbauer cells, APCs, and endothelial cells) as well as placental cell ligands that interact with decidual cell receptors (69). Accordingly, 91 sexually dimorphic receptor–ligand pairs were identified as being expressed by cell types at the maternal–fetal interface, which corresponded to processes such as immunomodulation, hormonal regulation, and metabolism (69). Thus, scRNA-seq can also provide evidence of sex-specific differences in cell–cell interactions that are characteristic of healthy pregnancies.

Single-cell landscape of RSA

Comparative studies have applied scRNA-seq technologies to investigate the cellular repertoire and interactions in obstetric disease during early pregnancy, namely RSA (71, 7375, 77). The single-cell investigation of decidual leukocyte subsets indicated an increased presence of T cells in RSA cases (73, 74), which was accompanied by diminished frequencies of specific macrophage (73, 74) and NK subsets (7375). For example, a CSF1+CD59+ NK cell subset prevalent in normal pregnancy was found to be reduced in RSA (75). Moreover, expression and interaction analyses indicated more active proinflammatory signaling for decidual leukocytes such as T cells, macrophages, and NK cells in RSA cases than in controls (7375). Macrophage interactions with T cells and NK cells are also altered in RSA since one macrophage subset showed enrichment of processes related to NK cell chemotaxis in healthy pregnancies but shifted toward processes related to T-cell chemotaxis in RSA (74). The latter finding was verified by using immunofluorescence staining, which revealed more prevalent colocalization of macrophages with NK cells in healthy pregnancy and more colocalization of macrophages with T cells in RSA (74). Thus, altered macrophage interactions with NK cells may result in aberrant promotion of T-cell responses associated with disease in early pregnancy (Fig. 1).

In addition to a general state of decidual immune activation, evaluation of the DSC compartment indicated a disease-driven shift in these cells, with several subsets diminishing in patients with RSA (71). Notably, a new DSC subset emerged only in RSA cases, which displayed gene expression enriched for cell apoptosis and senescence, potentially indicating abnormal DSC differentiation (71). Consistent with these observations, the developmental trajectories of DSCs were altered in RSA, with one potential differentiation pathway almost completely absent (71). Moreover, overall decidual cell–cell communications were increased in RSA cases (71). Cellular interaction analyses suggested that DSCs function as a central hub that communicates with other cell types in this compartment (71), thus defective DSC development or function could trigger a chain of adverse events implicated in RSA. This concept is further supported by the reported reduced expression of MAX (MYC-associated factor X) in DSCs from patients with RSA, which is implicated in proper decidualization and may indicate that impairment of this process is a contributing factor to disease development (77).

The above investigations demonstrate the capacity for single-cell technologies to provide novel insights into pregnancy diseases, such as RSA, which remain a challenge for obstetrics.

The second trimester represents a period of continued growth and development, characterized by a homeostatic environment at the maternal–fetal interface that is maintained by the resident cells (123126). One exploratory study of placental cell types included second-trimester samples as a means of comparing changes in single-cell composition throughout pregnancy (66). In this study, EVTs derived from the second-trimester placental villi formed two distinct clusters, one with enrichment of wound and cell adhesion processes and the other with enrichment of growth, response to stimulus, and hormone responses (66). Moreover, such EVTs represented the endpoint of the predicted differentiation pathway when combined with pseudotime trajectory analysis of first-trimester trophoblast subsets (66). These findings confirm that trophoblast differentiation continues throughout the second trimester. More recently, the developmental trajectories of CTBs were specifically explored in the human second-trimester placenta to avoid the inflammation and apoptosis associated with parturition in late pregnancy (127). In this study, both the smooth chorion (i.e., the chorion layer of the chorioamniotic membranes) and the villous chorion (within the placenta) were compared by using scRNA-seq (127). Consistent with other reports, major cell types included CTB, EVT, immune cells, stromal cells, and a small subset of epithelial cells (127). Notably, a subset of CTBs was identified as unique to the smooth chorion, termed SC-CTBs, and showed high expression of cytokeratins required for tissue integrity, as verified by immunofluorescence imaging (127). Trajectory analyses revealed that SC-CTBs in the smooth chorion and SCTs in the villous chorion shared a common progenitor (127). Notably, SC-CTBs exhibited greater inhibitory effects on EVTs than villous chorion CTBs in an ex vivo setting, and this effect was attributed to the SC-CTBs present in this compartment (127). Taken together, these findings point to distinct trophoblast cell types present in each compartment of the maternal–fetal interface that exhibit differential functionality despite originating from a shared progenitor.

In another recent investigation of the human maternal–fetal interface during the second trimester, researchers performed mass cytometry by time of flight to survey the leukocyte population present in this compartment (84). This technique was coupled with IMC and immunofluorescence staining to demonstrate the spatial distribution of specific cell types within the placental tissue as well as to validate tissue residence by localizing such cells outside of the fetal vasculature (84). CD4+ T cells and NK cells were abundant in the decidua, with macrophages more highly present in the placental villi (84). Specifically, the placental villi were enriched for subsets of CCR7 macrophages and DCs as well as CD8+CD69 T cells, suggesting a relatively inactive status of these cells (84). Deeper evaluation of expressed markers indicated subsets of CD163hi and CD163lo macrophages in the placental villi, with the former likely to be a classical Hofbauer subset, and evaluation of chemokine receptors on placental cells showed reduced expression of several of these markers, which was confirmed by the bulk RNA-seq data (84). The majority of placental T cells was found to be CD8+ memory T cells, with marker expression suggestive of Th2 differentiation, whereas CD4+ regulatory T cells (Tregs) were more abundant in the decidua (84). A later IMC-based study demonstrated similar findings and further indicated that the immune cell compartment undergoes changes in midpregnancy, such as a gradual increase in myeloid cells coupled with declining NK cell abundance (112). As noted above, such a decline in decidual NK cells included a substantial decrease in the abundance of the CD69+ dNK1 subset (112). Similarly, specific myeloid subsets showed distinct trends throughout gestation, supporting the concept that the overall changes in total NK cell or myeloid populations may not reflect the changes in individual subsets (112). Together, these observations provide an overview of the diverse immune cell landscape in the placental villi and provide further evidence of a largely homeostatic microenvironment at the maternal–fetal interface during midgestation.

Several investigations have also explored the murine placental tissues during midgestation to unravel the cellular differentiation taking place at this time (8183, 85). One study performed single-nucleus RNA sequencing to ensure adequate representation of SCTs and evaluated the murine placenta at days 9.5, 10.5, 12.5, and 14.5 of gestation (82). In this study, which focused on placental labyrinth development, the trophoblast population was dissected to identify sinusoidal trophoblast giant cells; SCT subsets including precursors, glycogen cells, spongiotrophoblasts, and their precursors; and junctional zone precursors (82). Such findings expanded on an earlier report that used scRNA-seq of the day 9.5 placenta to identify major clusters of progenitor trophoblasts and trophoblast giant cells in addition to DSCs, NK cells, and endothelial cells (83). Pseudotime trajectory analysis was implemented to describe several distinct differentiation pathways that included precursor, intermediate, and endpoint states as well as the key genes participating in each pathway (82). Moreover, based on the multiple sampled time points, additional temporal input was used to generate a map of placental development during midgestation and to infer the functional roles of each identified cell type in this process (82). In a separate investigation of the day 10.5 placenta, trajectory analyses were extended to include Hofbauer cell subsets, demonstrating a clear progression of monocyte to Hofbauer cluster 1 to Hofbauer cluster 2 (85). Consistent with the homeostatic microenvironment in early (32) and late (37) gestation, the most abundant placental cell–cell interactions at day 10.5 were between endothelial cell, pericyte, decidual cell, and labyrinth SCT pairs (85). Finally, another investigation focused on the fetal cells present in the placenta in midgestation by using a mating strategy wherein transgenic GFP+ male mice were mated with wild-type females (81). This study showed that hematopoietic precursors exist in the placenta that can give rise to a subset of fetal macrophages (81), shedding new light on cellular ontogeny in this organ.

Collectively, these studies elucidate the placental developmental processes taking place during midpregnancy and highlight this homeostatic period as an area for future investigations.

Placental cellular immunobiology

Single-cell technologies have allowed an unprecedented level of insight into the cellular populations and networks at the maternal–fetal interface in the third trimester, in the presence and absence of labor. Indeed, two pioneering studies focused on the cellular dynamics of the human placenta derived from term cesarean section deliveries to provide a single-cell atlas of this organ (78, 79). The first study focused on placental trophoblast cell types as well as a small subset of maternal immune cells and incorporated these data together with sequencing of SCTs collected by laser microdissection, primary undifferentiated endometrial stromal fibroblast cells, and primary decidual cells to obtain a combined dataset representative of placental cellular composition (78). The trophoblast clusters were assigned identities as VCT subsets and EVTs, with EVTs showing gene expression signatures associated with modulation of extracellular matrix, vascularization, and immune pathways (78). Interestingly, it was noted that SCTs displayed low-level expression of MHC-II, which is in contrast to other trophoblast subsets (78). The small maternal immune cell cluster was identified as DCs, which could be contaminating cells derived from the uterus (78). In a subsequent study, placental cell type clusters were identified that corresponded to populations of vascular endothelial cells, vascular smooth muscle cells, villous stromal cells, macrophages, trophoblasts, DSCs, DCs, T cells, and an erythrocytic subset, with the trophoblast cluster being further divided into EVTs, CTBs, and SCTs (79). By including paired biopsies sampled proximally and distally to the umbilical cord insertion point, it was shown that there is spatial heterogeneity reflected in the relative abundance of cell types such as DSCs and endothelial cells (79), which may be due to the convergence of chorionic arteries and veins at the umbilical cord (128). Analysis of the placental cell–cell communications network indicated likely interactions between adjacent maternal and placental cell types, with DSCs showing a high amount of signaling to and from SCTs and EVTs (78). Intertrophoblast communication was also highlighted since each CTB subset displayed putative interactions with SCTs and EVTs (78). Notably, by contrasting cell–cell communications using the undifferentiated endometrial stromal fibroblast cells and DSCs, it was found that decidualization enhances signaling potential between DSCs and the fetal trophoblast (78). Together, these two pioneer studies provided important new insights into placental cellular interactions as well as valuable single-cell datasets that were leveraged by later investigations (80, 129131).

To enhance the translational value of single-cell placental dynamics, Tsang et al. extrapolated the cellular signatures to the maternal circulating cell-free RNA (79). This novel method was based on studies showing that cell-free DNA and RNA derived from the fetus/placenta are found in the maternal circulation (132139). Using this approach, it was found that expression profiles corresponding to DSCs, endothelial cells, smooth muscle cells, stromal cells, and monocytes steadily increased in the maternal circulation throughout pregnancy (79). By contrast, EVT, SCT, and B-cell profiles tended to decrease, particularly toward the end of gestation (79). Finally, the overall T-cell population signature decreased in midpregnancy and then increased in the third trimester and postpartum period (79), which is in line with the concept that T-cell activation is implicated in the inflammatory milieu that accompanies term parturition at the maternal–fetal interface (3840, 43, 45, 50, 140).

A recent report focused on placental mesenchymal stem/stromal cells demonstrated multiple subsets contained within this population, including some with immunomodulatory gene expression signatures (141). Moreover, these cells expressed cytokines such as CCL2, immunomodulatory factors such as IFN-induced transmembrane proteins and the regulatory factor PRDM1 (BLIMP-1), and a single-cell assay for transposase-accessible chromatin with high-throughput sequencing indicated high chromatin accessibility of immune regions (141), further supporting the involvement of placental mesenchymal stem/stromal cells in immune processes. Moreover, the findings generated in this study support the potential application of placental mesenchymal stem/stromal cells for regenerative or immunomodulatory cell-based therapies.

To date, only one report has provided comparative scRNA-seq analysis of the human placental villi in the presence and absence of spontaneous labor at term (80). In particular, relative differences in the proportions of CTB subsets, activated T cells, monocytes, and macrophages are observed between the labor and nonlabor placental villi, with such immune subsets including cells of maternal and fetal origin (80). Moreover, term labor was associated with substantial changes in gene expression across multiple placental cell types, including macrophages, monocytes, stromal cells, EVTs, and CTBs (80). Such expression changes were enriched for labor-associated terms, such as vascular smooth muscle contraction in the fibroblast subset and cell cycle/metabolism in EVTs (80). Moreover, by applying an approach similar to that reported in (79), gene signatures corresponding to placental cell types could be monitored in the maternal circulation throughout pregnancy, and signatures of NK cells and T cells were found to be enhanced with term labor (80). Thus, such analyses provide not only insight into the labor-specific placental changes that occur at the single-cell level but also a potential means of monitoring pregnancy and labor in the maternal circulation.

Decidual cellular immunology

The cellular composition of the decidua undergoes modification throughout gestation in preparation for the inflammatory process of labor (45, 47, 140, 142). Single-cell surveys of the decidua obtained from term cesarean deliveries characterized the overall proportions of cell types in this compartment, including a substantial fraction of T cells (both resting and activated), NK cells, DSCs, endothelial cells, and fibroblasts as well as invasive EVTs, and subsets such as macrophages showed substantial labor-associated changes in gene expression (80, 87) (Fig. 2). These data are in line with prior cytological surveys of the decidua showing that leukocytes are attracted to this compartment before the onset of labor (40, 43, 49, 140, 143146). Moreover, a new subset of lymphatic endothelial decidual (LED) cells described in the chorioamniotic membranes displayed an expression profile enriched for cellular interactions and adhesion (80). This observation suggested that LED cells present in the chorioamniotic membranes may be functionally mediating the influx of immune cells into this compartment during the process of labor (80). Indeed, immunofluorescence staining of the chorioamniotic membranes revealed the coexpression of the lymphatic marker LYVE1 and the endothelial marker CD31, demonstrating the presence of lymphatic vessels in the decidua parietalis (80). Consistently, a single-cell study focused on decidual endothelial subsets in term nonlabor deliveries identified five distinct cell clusters with differing expression profiles, two of which were enriched for cell adhesion processes (86). Together, these studies suggested that decidual endothelial cells, including the novel LED cell type, can contribute to the accumulation of infiltrating lymphocytes into the maternal–fetal interface in preparation for and during term parturition.

Mucosal-associated invariant T (MAIT) cells, an innate-like T-cell subset expressing a restricted TCR (147), have also been detected at the maternal–fetal interface (148150); yet, the expression profiles and functionality of these cells has been underexplored. A targeted investigation used combined flow cytometry, cellular indexing of transcriptomes and epitopes by sequencing, and scRNA-seq approaches to characterize MAIT cells in the decidua basalis and decidua parietalis, demonstrating that the majority of these cells displayed an effector memory phenotype (151). Decidual MAIT cells exhibited gene expression profiles distinct from those found in the periphery, including genes involved in immune suppression and cell migration (151). Key differentiation factors upregulated in MAIT cells compared to conventional T cells included PLZF and EOMES, and in vitro stimulation resulted in secretion of IFN-γ and TNF together with granzyme B, indicating that these cells are functional and can participate in maternal-fetal immune responses (151).

Recent investigations have also compared the changes in cellular composition and cell–cell signaling between decidual tissues derived from labor and nonlabor deliveries to evaluate the participation of individual subsets in the inflammatory process of parturition. Spontaneous term labor was shown to result in the increased prevalence of activated T cells, monocytes, macrophages, and DSCs in the decidua basalis compared to term deliveries without labor (80). Moreover, IMC analysis of the maternal–fetal interface at term indicated increased abundance of myeloid cells and T cells compared to second-trimester samples (112). Such changes are consistent with prior studies indicating an influx of immune cells to the maternal–fetal interface during parturition (40, 43, 47, 140, 143, 145).

More prominent than changes in cell-type proportions are the transcriptomic changes that occur to facilitate labor. Macrophages, stromal cells, monocytes, T cells, and EVTs, among others, showed drastic differential gene expression during term labor compared to their nonlabor counterparts, and such genes were enriched for inflammatory and immunomodulatory pathways (80). Similarly, each decidual endothelial cell cluster showed labor-specific enrichment of inflammatory pathways such as IL-17 signaling, leukocyte differentiation, myeloid differentiation, and cytokine-related terms (86), as did DSC and decidual EVT subsets (87), suggesting that decidual immune and tissue cell subsets propagate inflammation to drive parturition. Such changes in the transcriptomic profiles of decidual cells manifest in altered cell–cell communications, as indicated by numerous enhanced signaling pathways such as IL-1β/IL-1R1, IL-6/IL-6R, TNF superfamily member 14/lymphotoxin-β receptor, and multiple chemokine/chemokine receptor pathways (88). Rather than solely regulating new pathways, labor also involves the continued upregulation or downregulation of already modulated signaling pathways (88), suggesting that transcriptomic changes implicated in labor may be initiated earlier in gestation. In particular, T cells undergo significant shifts in intercellular communications with other decidual cells, involving pathways such as Notch, NF-κB, MAPK, JAK/STAT, and chemokine signaling, among others (88). However, these underlying labor-associated transcriptomic changes did not seem to impact the TCR clonality and diversity within the decidua (88), consistent with the concept that the decidual T-cell compartment is composed primarily of memory T cells (43, 84, 151). Together, these data point to a decidual inflammatory response implicated in labor that involves transcriptomic changes in specific cell types from the innate and adaptive limbs of the immune system.

The use of scRNA-seq to decipher the mechanisms leading to preterm labor

Spontaneous preterm labor is not simply the premature activation of the common pathway of parturition but rather represents a pathological process distinct from normal labor at term (152). Unraveling the molecular mechanisms underlying spontaneous preterm labor is therefore an ongoing endeavor within our group. Thus, we have leveraged scRNA-seq to compare and contrast the cellular composition and transcriptomic profiles of the decidua, placenta, and chorioamniotic membranes in term and preterm labor to improve the understanding and potential prediction of this obstetrical syndrome (80) (Fig. 2). Compared to term labor, preterm labor involves substantial changes in gene expression within the EVT and CTB populations, potentially indicating a distinct response in these cell types (80). Moreover, within preterm labor-affected subsets such as EVTs and CTBs, specific genes were identified that showed an opposite direction of change compared to term labor, indicating that distinct cellular mechanisms are involved in these processes (80). To extend the clinical relevance of our findings, we applied our scRNA-seq–derived signatures to the cellular transcriptome of the maternal circulation and showed that signatures of maternal macrophages, monocytes, and activated T cells were elevated in women who underwent spontaneous preterm labor compared to gestational age-matched control subjects, supporting a role for intravascular immune activation in the pathophysiology of preterm labor (80). Thus, the maternal circulation may provide a useful window that can be used to monitor preterm labor-specific events at the maternal–fetal interface.

More recently, we undertook the investigation of the cellular interactions implicated in pathological labor by using a murine model of preterm birth induced by the intra-amniotic inoculation of Escherichia coli (90). We evaluated the key tissues implicated in the common pathway of labor (i.e., the uterus, decidua, and cervix) at single-cell resolution and demonstrated that preterm labor affects the cellularity of the uterus, decidua, and cervix through immune cell infiltration and altered transcriptomic profiles of nonimmune cell types in a tissue-specific manner (90). In the uterus, both innate and adaptive immune cell subsets (e.g., neutrophils, macrophages, DCs, NK cells, and T cells) contributed to pathways implicated in preterm labor, such as cytokine and chemokine signaling, as did nonimmune subsets (e.g., fibroblasts, stromal cells, epithelial cells, smooth muscle cells, and endothelial cells), and the interaction strength among cell types was altered with preterm labor (90). Similar changes in cell–cell signaling were observed in the decidua; yet, unique pathways such as IL-17 were also revealed in this tissue (90). The cellularity of the cervix predominantly involved epithelial cells, which showed increased incoming interactions from other tissue cell subsets during preterm labor (90). Importantly, although specific cell types displayed consistent signaling across tissues, each also exhibited tissue-specific processes with preterm labor, indicating unique functions tailored to the tissue microenvironment (90). Together, these findings provide novel insight into the cellular changes taking place in the murine reproductive tissues during preterm labor and lead to premature delivery.

Placental scRNA-seq sheds light into the pathophysiology of preeclampsia

Preeclampsia is primarily a placental and intravascular disease (153), thus this tissue has been the target of single-cell investigations aimed at uncovering the molecular mechanisms underlying the development of this obstetrical syndrome (79, 154156). One of the first comprehensive scRNA-seq studies integrated placenta-derived single-cell signatures with bulk transcriptomic data derived from the plasma cell–free RNA of women with early-onset preeclampsia and healthy pregnancies (79). Signatures corresponding to decidual cells, endothelial cells, and EVTs were upregulated in the maternal plasma with preeclampsia, whereas the SCT signature was reduced (79). Analysis of the transcriptional heterogeneity of genes involved in cell migration, cell proliferation, and apoptosis were more variable in patients with preeclampsia than in those with a normal term delivery; moreover, genes annotated to cell death had overall higher expression in preeclampsia (79), which is in tandem with prior associations between this disease and trophoblast apoptosis (157161) (Fig. 2). Consistently, the direct scRNA-seq comparison of preeclamptic and healthy placentas indicated disease-driven enrichment of genes annotated to oxidative stress and inflammation in EVTs (154, 156) and those annotated to cell cycle and protein folding in SCTs (156). Moreover, clustering of differentially regulated genes revealed a module with reduced activity in preeclampsia related to cytokines, regulation of cell death, and differentiation (156). Two transcription factors, CEBPB and GTF2B, were identified as being greatly reduced in the disease state and thus could be implicated in the trophoblast dysfunction characteristic of preeclampsia (156).

Preeclampsia has traditionally been classified into two subsets, with each being characterized by distinct pathophysiology: early-onset and late-onset (153, 162164). To unravel cellular changes that may distinguish early- and late-onset preeclampsia, an in silico investigation used placental single-cell signatures derived from the study of Vento-Tormo et al. (68) to deconvolute bulk transcriptomic data of placentas from women with either form of the disease (155). The number of genes differentially regulated in early-onset preeclampsia compared with its control group was substantially greater than in late-onset preeclampsia (155), which is consistent with early-onset preeclampsia being largely a placental disease (153, 165167). Moreover, the direct comparison of early- and late-onset preeclampsia also resulted in a large set of differentially expressed genes, further indicating distinct underlying pathophysiology (155). Deeper investigation of changes at single-cell resolution indicated that EVTs and fibroblasts play a key role in early-onset preeclampsia, displaying dysregulated signaling associated with angiogenesis and fibrosis (155). Importantly, novel trophoblast-derived markers, such as EBI3 (EBV-induced 3), were identified as upregulated in early-onset preeclampsia, and such proteins could be detected as elevated in the maternal circulation (155). Therefore, the combination of placental single-cell transcriptomics with soluble biomarker discovery can represent a viable approach for the prediction and diagnosis of preeclampsia.

Myometrial cell types and their contributions to spontaneous term labor

The uterine myometrium and the cervix are key organs in the common pathway of parturition (168170). Thus, investigation of the cellular processes taking place in these tissues before the initiation of labor can provide a baseline measurement useful for the study of physiological or pathological labor. A recent study obtained myometrial and cervical biopsies during planned cesarean hysterectomies without evidence of labor in the third trimester and performed scRNA-seq (171). The resulting data were cross-referenced with the Human Protein Atlas to identify major cell-type clusters, which included endothelial, epithelial, and stromal cells; smooth muscle cells; and leukocytes (171). Expression of CD74 (HLA class II histocompatibility Ag γ-chain) could distinguish the epithelial, endothelial, and leukocyte clusters from the stromal and smooth muscle cells (171). Because CD74 regulates the function of MHC-II molecules (172) and is thought to be restricted mainly to APCs under homeostatic conditions, a potential underlying state of tissue inflammation was proposed that could drive CD74 expression in epithelial/endothelial cells (173, 174). This finding was consistent with the observed upregulation of genes associated with inflammation (171); yet, given that biopsies were taken before active labor, such inflammation could represent preparation for the onset of parturition.

We recently undertook a single-cell survey of the human uterine tissue (primarily the myometrium) to further investigate the molecular mechanisms underlying the transition from a quiescent to a contractile state before term labor (89). Myometrial biopsies were obtained from term deliveries either with or without spontaneous labor for scRNA-seq, from which a total of 24 immune and nonimmune cell types were identified, including multiple subsets of smooth muscle cells and macrophages (89). Specifically, we classified three subsets of smooth muscle cells according to their transcriptomic profiles, which were then validated by protein expression (89). We showed that the first smooth muscle subset displayed an expression profile involving smooth muscle contraction, the second included processes related to neutrophil biology (e.g., neutrophil elastase expression), and the third exhibited increased IFN-γ signaling, demonstrating the differing roles of these distinct subsets in the process of labor (89). Transcriptomic characterization of macrophage subsets and stromal cell types was also reported (89). Comparison of overall changes in cell abundance indicated a substantial labor-specific increase in the presence of stromal cells, endothelial cells, monocytes, decidual cells, and myofibroblasts, which was accompanied by reductions in macrophage and lymphocyte subsets (89). Consistently, the most dramatic labor-driven changes in gene expression were found in stromal cells, endothelial cells, monocytes, and macrophages, which included an overall enrichment of multiple muscle- and contraction-related processes (89). We also evaluated intercellular communications and identified major signaling pathways implicated in labor, such as complement, contraction, IL-1, TGF-β, and THY1 (89). In particular, we reported that specific myometrial cell types, such as decidual cells, EVTs, myofibroblasts, smooth muscle cells, and stromal cells, act as receivers of IL-1 (89), which is considered a master regulator of human parturition (175178). By comparing scRNA-seq data with bulk transcriptomics of the human myometrium, we demonstrated a high degree of agreement between these datasets, with single-cell technology providing better coverage of differential gene expression (89). Consistent with prior analyses, gene expression signatures corresponding to scRNA-seq–derived myometrial cell types could be monitored in the maternal circulation, allowing evaluation of cellular dynamics throughout pregnancy (89). Importantly, such comparison indicated agreement in the labor-specific enrichment of specific cell-type signatures, namely monocytes, thereby providing potential biomarkers that could be indicative of labor progression (89).

In addition to providing insight into the cellular changes and interactions associated with term labor, single-cell technology has also been applied to evaluate resident regulatory T cells (Tregs) present in the placental bed (uterine tissues) biopsied after term cesarean delivery (179). Uterine Tregs (uTregs) expressed a core transcriptomic signature consistent with classical Tregs, which was even more pronounced than peripheral Tregs and included expression of FOXP3, CTLA4, and IL2RA (179). In vitro assays confirmed the suppressive functionality of these cells, consistent with the observed gene expression/pathway enrichment indicative of activated, effector uTregs (179). By comparing uTregs to publicly available Treg datasets from other tissues, it was shown that such cells displayed transcriptomic overlap with tissue- and tumor-infiltrating Tregs (179). These data provide invaluable insight into uTregs that participate in the modulation of local effector T cells to prevent aberrant immune activation during pregnancy.

Together, the studies outlined above point to the myometrium as an underexplored site of cellular interactions and signaling necessary for physiological labor at term. Future single-cell investigations may further explore such pathways in the context of pathological labor leading to preterm birth as well as other pregnancy complications.

Single-cell profile of the maternal–fetal interface in patients with COVID-19

Since the emergence of SARS-CoV-2 in late 2019, investigators around the globe have shifted research efforts to uncover the molecular mechanisms that dictate maternal infection, disease severity, and risk of vertical transmission during pregnancy. Initially, to further this goal, multiple reports undertook in silico analysis of previously generated single-cell data (66, 68, 78) to evaluate the expression of the canonical SARS-CoV-2 cell entry mediators ACE2 (angiotensin-converting enzyme 2) (180182) and TMPRSS2 (transmembrane protease serine 2) (182) at the maternal–fetal interface (92, 9496, 99, 183, 184). Each of these studies observed independent expression of ACE2 and TMPRSS2 in decidual cells, such as DSCs and perivascular cells (92, 96, 183), and/or in placental trophoblast subsets (92, 95, 96, 184). However, viral cell entry requires the coexpression of both ACE2 and TMPRSS2 within the same cell (182), and the evidence for coexpression of such cell entry mediators by placental cells was unclear, given that some studies indicated detectable coexpression (95, 184), whereas others suggested this was rare (94, 96, 99). Therefore, to provide further clarity in this regard, we undertook a combined approach that used (1) previous single-cell data of the first-trimester maternal–fetal interface (68), (2) new single-cell data of the decidua and placenta from an indicated second-trimester hysterectomy, and (3) our previous single-cell dataset of the third-trimester decidua basalis, placental villi, and chorioamniotic membranes (80, 91). We found that a minimal number of cells coexpress ACE2 and TMPRSS2 at the maternal–fetal interface throughout gestation, even with an extremely permissive expression threshold (91). To overcome the limitation of low SCT representation in our single-cell dataset (due to their multinucleated morphology), single-nucleus RNA sequencing of placental tissues was performed, which confirmed that coexpression of ACE2 and TMPRSS2 is minimal among these cells (91). Thus, our data support the absence of meaningful coexpression of classical cell entry mediators for SARS-CoV-2 at the maternal–fetal interface throughout pregnancy, a concept that has since been further confirmed (185).

Regardless of the expression patterns of viral cell entry mediators, a central question amid the COVID-19 pandemic has been whether maternal infection results in a fetal-placental immune response that can lead to adverse pregnancy outcomes and, more important, whether the fetus itself is impacted. A study used bulk and single-cell transcriptomic analyses of the placental tissues from women diagnosed with COVID-19 to demonstrate upregulation of genes associated with immune response compared to uninfected control subjects (93). At single-cell resolution, such COVID-19–driven changes included enrichment of cytotoxic molecules in NK cells and signs of T-cell activation, and endothelial cells similarly displayed signs of activation and immune response (93). This finding is consistent with a case report in which scRNA-seq was performed using placental tissues from a pregnant patient with COVID-19 who was delivered at 28 wk of gestation, showing that CD8+ T cells were activated (98). Moreover, cell–cell communication analysis indicated increased interactions between immune cells at the maternal–fetal interface in COVID-19 (93, 98), including between T cells and monocytes/NK cells (93). In light of this evidence pointing to a maternal immune response induced by COVID-19 during pregnancy, we undertook a comprehensive multidisciplinary investigation of maternal-fetal immunity in pregnant women with SARS-CoV-2 infection, most of whom were asymptomatic, and used scRNA-seq to show that maternal T cells and macrophages in the chorioamniotic membranes display substantial changes in gene expression compared to cells from uninfected pregnant women (97). We then compared our T-cell signatures to previously generated single-cell signatures of peripheral T cells from hospitalized patients with COVID-19, and we observed a positive correlation between these datasets (97), suggesting that T cells derived from the maternal–fetal interface of pregnant patients with COVID-19 display characteristics similar to those found in the circulation of nonpregnant patients with severe disease. Shared genes were enriched for protein translation processes; yet, some differentially expressed genes were unique to maternal–fetal interface-derived T cells (97). The combined differentially expressed genes from the maternal–fetal interface of women with COVID-19 showed enrichment of multiple IFN signaling pathways, indicating that SARS-CoV-2 infection drives an antiviral immune response even when the virus itself is not present in this compartment (97). Overall, these novel findings support the value of single-cell datasets that can be leveraged to investigate relevant cellular and molecular targets for diseases such as COVID-19, which, in turn, can inform translational research directed at therapeutic interventions.

Limitations of single-cell technology

The emergence and popularization of single-cell techniques has opened new avenues of scientific discovery across multiple disciplines. Yet, the widespread use of such methods has also highlighted their limitations in comparison to other cellular and molecular tools, such as the inherent discovery-based nature of current single-cell approaches. Moreover, the analysis of scRNA-seq datasets requires advanced computational approaches that rely on algorithmic clustering and associations to determine cell identities and profiles, which can differ on the basis of the platforms and analysis pipelines used (as has been reviewed extensively in [186189] and elsewhere). For this reason, researchers who utilize single-cell approaches have often chosen to validate key findings (e.g., identification of novel cell subsets) with alternative approaches such as flow cytometry, imaging methods, or animal models. Indeed, we have used immunofluorescence imaging to confirm our identification of a potentially novel cell type, LED cells, in the human chorioamniotic membranes (80). Similarly, we used the same approach to validate our identification of distinct subsets of smooth muscle cells in the human myometrium based on expression of oxytocin receptor, elastase, or IFN-γ (89). For studies in which the identification of novel cell types represents a primary outcome, we consider that additional functional and phenotypic analyses are essential to ensure that such populations do not represent artifacts resulting from data processing and analysis. Overall, we consider that the application of validation techniques is essential for providing confirmatory analysis of discoveries made by using exploratory single-cell approaches.

An important consideration for scRNA-seq and other single-cell technologies is the analytical approach that will be used to dissect the generated data. A complete discussion of single-cell analysis pipelines and tools is outside the scope and expertise of this review, and such topics have been covered more extensively elsewhere (186, 187, 189). Yet, we have successfully applied several of the tools described in this review, such as CellChat cell–cell communication analysis (118) and Slingshot trajectory analysis (190), to our single-cell investigations of the human maternal–fetal interface (Fig. 3). In particular, an important consideration when studying the placental and decidual tissues at single-cell resolution is the maternal or fetal origin of each cell. To overcome this potential limitation and aid in the interpretation of our scRNA-seq data, we have successfully incorporated maternal and fetal genotyping data into our analysis pipeline (80, 89, 97, 191). Other limitations of scRNA-seq in the context of maternal-fetal immunology include the difficulty of obtaining biopsies for research, for which we have proposed and used two different translational solutions. First, we have shown that specific cellular signaling pathways implicated in labor are shared between the human and murine myometrium (90). Although validation remains necessary when comparing humans and mice, such findings can lay the groundwork for future single-cell investigations of pregnancy. In parallel, the evaluation of placenta-derived single-cell signatures in the maternal circulation has been shown to represent a viable approach for monitoring pregnancy and its complications in a noninvasive manner, as described in this review (79, 80, 89, 130). Taken together, these findings indicate that scRNA-seq represents a translationally relevant approach for investigation of the maternal–fetal interface; yet, we consider that the careful application of this technology and its analysis is required to ensure the generation of useful and reproducible results.

Collectively, the reports summarized herein emphasize the value of single-cell technology for characterizing the cellular dynamics and interactions at the maternal–fetal interface. Given the cellular heterogeneity of the decidua, placenta, and reproductive tissues, “omics” techniques that provide information at single-cell resolution are rapidly becoming the new standard for investigations of the molecular mechanisms underlying developmental processes, physiological and pathological labor, and other obstetrical diseases. The findings reviewed herein provide a rich framework of single-cell data, generated at different gestational time points and under various conditions, that can be a starting point for future studies. Importantly, we recognize that there are often disparities between studies which can result from differences in technologies, individual platforms, and cell-type classification methods, among other variables. Thus, given the abundance of single-cell datasets that have been generated from the maternal-fetal unit, there is potential for unified analyses that integrate such data under consistent cell-type annotations and analysis pipelines to represent single-cell dynamics at different time points throughout pregnancy in a comparable manner. In summary, this review demonstrates the power of single-cell technologies to uncover new cellular immunobiological pathways and interactions that may have translational relevance for obstetric disease, and it is our hope that this summary can act as a catalyst for future investigations of the maternal–fetal interface.

This work was supported by the Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services under Contract No. HHSN275201300006C. This work was also supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health. R.R. has contributed to this work as part of his official duties as an employee of the U.S. federal government. The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Abbreviations used in this article:

CTB

cytotrophoblast

DC

dendritic cell

DSC

decidual stromal cell

EVT

extravillous trophoblast

IMC

imaging mass cytometry

LED

lymphatic endothelial decidual

MAIT

mucosal-associated invariant T

MHC-II

MHC class II

RSA

recurrent spontaneous abortion

SC-CTB

cytotrophoblast unique to the smooth chorion

scRNA-seq

single-cell RNA sequencing

SCT

syncytiotrophoblast (termed “STB” in some studies)

Treg

regulatory T cell

uTreg

uterine regulatory T cell

VCT

villous cytotrophoblast

1.
Cunningham
F. G.
,
K. J.
Leveno
,
J. S.
Dashe
,
B. L.
Hoffman
,
C. Y.
Spong
,
B. M.
Casey
.
2022
.
Embryogenesis and fetal development.
In
Williams Obstetrics
, 26th Ed.
McGraw Hill
,
New York, NY
.
2.
Norwitz
E. R.
,
D. J.
Schust
,
S. J.
Fisher
.
2001
.
Implantation and the survival of early pregnancy.
N. Engl. J. Med.
345
:
1400
1408
.
3.
Red-Horse
K.
,
Y.
Zhou
,
O.
Genbacev
,
A.
Prakobphol
,
R.
Foulk
,
M.
McMaster
,
S. J.
Fisher
.
2004
.
Trophoblast differentiation during embryo implantation and formation of the maternal-fetal interface.
J. Clin. Invest.
114
:
744
754
.
4.
Moffett
A.
,
C.
Loke
.
2006
.
Immunology of placentation in eutherian mammals.
Nat. Rev. Immunol.
6
:
584
594
.
5.
Cha
J.
,
X.
Sun
,
S. K.
Dey
.
2012
.
Mechanisms of implantation: strategies for successful pregnancy.
Nat. Med.
18
:
1754
1767
.
6.
Arck
P. C.
,
K.
Hecher
.
2013
.
Fetomaternal immune cross-talk and its consequences for maternal and offspring’s health.
Nat. Med.
19
:
548
556
.
7.
Lash
G. E.
2015
.
Molecular cross-talk at the feto-maternal interface.
Cold Spring Harb. Perspect. Med.
5
:
a023010
.
8.
Griffith
O. W.
,
A. R.
Chavan
,
S.
Protopapas
,
J.
Maziarz
,
R.
Romero
,
G. P.
Wagner
.
2017
.
Embryo implantation evolved from an ancestral inflammatory attachment reaction.
Proc. Natl. Acad. Sci. USA
114
:
E6566
E6575
.
9.
Ander
S. E.
,
M. S.
Diamond
,
C. B.
Coyne
.
2019
.
Immune responses at the maternal-fetal interface.
Sci. Immunol.
4
:
eaat6114
.
10.
Petroff
M. G.
2005
.
Immune interactions at the maternal-fetal interface.
J. Reprod. Immunol.
68
:
1
13
.
11.
Sargent
I. L.
,
A. M.
Borzychowski
,
C. W.
Redman
.
2006
.
NK cells and human pregnancy – an inflammatory view.
Trends Immunol.
27
:
399
404
.
12.
Erlebacher
A.
2013
.
Immunology of the maternal-fetal interface.
Annu. Rev. Immunol.
31
:
387
411
.
13.
Mori
M.
,
A.
Bogdan
,
T.
Balassa
,
T.
Csabai
,
J.
Szekeres-Bartho
.
2016
.
The decidua—the maternal bed embracing the embryo—maintains the pregnancy.
Semin. Immunopathol.
38
:
635
649
.
14.
Tersigni
C.
,
C. W.
Redman
,
R.
Dragovic
,
D.
Tannetta
,
G.
Scambia
,
N.
Di Simone
,
I.
Sargent
,
M.
Vatish
.
2018
.
HLA-DR is aberrantly expressed at feto-maternal interface in pre-eclampsia.
J. Reprod. Immunol.
129
:
48
52
.
15.
Holder
B.
,
J. D.
Aplin
,
N.
Gomez-Lopez
,
A. E. P.
Heazell
,
J. L.
James
,
C. J. P.
Jones
,
H.
Jones
,
R. M.
Lewis
,
G.
Mor
,
C. T.
Roberts
, et al
2021
.
‘Fetal side’ of the placenta: anatomical mis-annotation of carbon particle ‘transfer’ across the human placenta.
Nat. Commun.
12
:
7049
.
16.
Gellersen
B.
,
I. A.
Brosens
,
J. J.
Brosens
.
2007
.
Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives.
Semin. Reprod. Med.
25
:
445
453
.
17.
Shima
T.
,
Y.
Sasaki
,
M.
Itoh
,
A.
Nakashima
,
N.
Ishii
,
K.
Sugamura
,
S.
Saito
.
2010
.
Regulatory T cells are necessary for implantation and maintenance of early pregnancy but not late pregnancy in allogeneic mice.
J. Reprod. Immunol.
85
:
121
129
.
18.
Chen
T.
,
G.
Darrasse-Jèze
,
A. S.
Bergot
,
T.
Courau
,
G.
Churlaud
,
K.
Valdivia
,
J. L.
Strominger
,
M. G.
Ruocco
,
G.
Chaouat
,
D.
Klatzmann
.
2013
.
Self-specific memory regulatory T cells protect embryos at implantation in mice.
J. Immunol.
191
:
2273
2281
.
19.
Shima
T.
,
K.
Inada
,
A.
Nakashima
,
A.
Ushijima
,
M.
Ito
,
O.
Yoshino
,
S.
Saito
.
2015
.
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.
J. Reprod. Immunol.
108
:
72
82
.
20.
Robertson
S. A.
,
A. S.
Care
,
L. M.
Moldenhauer
.
2018
.
Regulatory T cells in embryo implantation and the immune response to pregnancy.
J. Clin. Invest.
128
:
4224
4235
.
21.
Schumacher
A.
,
A. C.
Zenclussen
.
2019
.
Human chorionic gonadotropin-mediated immune responses that facilitate embryo implantation and placentation.
Front. Immunol.
10
:
2896
.
22.
Hofmann
A. P.
,
S. A.
Gerber
,
B. A.
Croy
.
2014
.
Uterine natural killer cells pace early development of mouse decidua basalis.
Mol. Hum. Reprod.
20
:
66
76
.
23.
Yang
F.
,
Q.
Zheng
,
L.
Jin
.
2019
.
Dynamic function and composition changes of immune cells during normal and pathological pregnancy at the maternal-fetal interface.
Front. Immunol.
10
:
2317
.
24.
Chaouat
G.
,
G. A.
Voisin
,
D.
Escalier
,
P.
Robert
.
1979
.
Facilitation reaction (enhancing antibodies and suppressor cells) and rejection reaction (sensitized cells) from the mother to the paternal antigens of the conceptus.
Clin. Exp. Immunol.
35
:
13
24
.
25.
Bonney
E. A.
,
J.
Onyekwuluje
.
2003
.
The H-Y response in mid-gestation and long after delivery in mice primed before pregnancy.
Immunol. Invest.
32
:
71
81
.
26.
Aluvihare
V. R.
,
M.
Kallikourdis
,
A. G.
Betz
.
2004
.
Regulatory T cells mediate maternal tolerance to the fetus.
Nat. Immunol.
5
:
266
271
.
27.
Zenclussen
A. C.
,
K.
Gerlof
,
M. L.
Zenclussen
,
A.
Sollwedel
,
A. Z.
Bertoja
,
T.
Ritter
,
K.
Kotsch
,
J.
Leber
,
H. D.
Volk
.
2005
.
Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model.
Am. J. Pathol.
166
:
811
822
.
28.
Robertson
S. A.
,
L. R.
Guerin
,
L. M.
Moldenhauer
,
J. D.
Hayball
.
2009
.
Activating T regulatory cells for tolerance in early pregnancy — the contribution of seminal fluid.
J. Reprod. Immunol.
83
:
109
116
.
29.
Kahn
D. A.
,
D.
Baltimore
.
2010
.
Pregnancy induces a fetal antigen-specific maternal T regulatory cell response that contributes to tolerance.
Proc. Natl. Acad. Sci. USA
107
:
9299
9304
.
30.
Rowe
J. H.
,
J. M.
Ertelt
,
L.
Xin
,
S. S.
Way
.
2012
.
Pregnancy imprints regulatory memory that sustains anergy to fetal antigen.
Nature
490
:
102
106
.
31.
Samstein
R. M.
,
S. Z.
Josefowicz
,
A.
Arvey
,
P. M.
Treuting
,
A. Y.
Rudensky
.
2012
.
Extrathymic generation of regulatory T cells in placental mammals mitigates maternal-fetal conflict.
Cell
150
:
29
38
.
32.
Care
A. S.
,
K. R.
Diener
,
M. J.
Jasper
,
H. M.
Brown
,
W. V.
Ingman
,
S. A.
Robertson
.
2013
.
Macrophages regulate corpus luteum development during embryo implantation in mice.
J. Clin. Invest.
123
:
3472
3487
.
33.
Jiang
T. T.
,
V.
Chaturvedi
,
J. M.
Ertelt
,
J. M.
Kinder
,
D. R.
Clark
,
A. M.
Valent
,
L.
Xin
,
S. S.
Way
.
2014
.
Regulatory T cells: new keys for further unlocking the enigma of fetal tolerance and pregnancy complications.
J. Immunol.
192
:
4949
4956
.
34.
Svensson-Arvelund
J.
,
R. B.
Mehta
,
R.
Lindau
,
E.
Mirrasekhian
,
H.
Rodriguez-Martinez
,
G.
Berg
,
G. E.
Lash
,
M. C.
Jenmalm
,
J.
Ernerudh
.
2015
.
The human fetal placenta promotes tolerance against the semiallogeneic fetus by inducing regulatory T cells and homeostatic M2 macrophages.
J. Immunol.
194
:
1534
1544
.
35.
Bonney
E. A.
2016
.
Immune regulation in pregnancy: a matter of perspective?
Obstet. Gynecol. Clin. North Am.
43
:
679
698
.
36.
Gomez-Lopez
N.
,
M.
Arenas-Hernandez
,
R.
Romero
,
D.
Miller
,
V.
Garcia-Flores
,
Y.
Leng
,
Y.
Xu
,
J.
Galaz
,
S. S.
Hassan
,
C. D.
Hsu
, et al
2020
.
Regulatory T cells play a role in a subset of idiopathic preterm labor/birth and adverse neonatal outcomes.
Cell Rep.
32
:
107874
.
37.
Gomez-Lopez
N.
,
V.
Garcia-Flores
,
P. Y.
Chin
,
H. M.
Groome
,
M. T.
Bijland
,
K. R.
Diener
,
R.
Romero
,
S. A.
Robertson
.
2021
.
Macrophages exert homeostatic actions in pregnancy to protect against preterm birth and fetal inflammatory injury.
JCI Insight
6
:
e146089
.
38.
Sindram-Trujillo
A. P.
,
S. A.
Scherjon
,
P. P.
van Hulst-van Miert
,
H. H.
Kanhai
,
D. L.
Roelen
,
F. H.
Claas
.
2004
.
Comparison of decidual leukocytes following spontaneous vaginal delivery and elective cesarean section in uncomplicated human term pregnancy.
J. Reprod. Immunol.
62
:
125
137
.
39.
Osman
I.
,
A.
Young
,
F.
Jordan
,
I. A.
Greer
,
J. E.
Norman
.
2006
.
Leukocyte density and proinflammatory mediator expression in regional human fetal membranes and decidua before and during labor at term.
J. Soc. Gynecol. Investig.
13
:
97
103
.
40.
Gomez-Lopez
N.
,
G.
Estrada-Gutierrez
,
L.
Jimenez-Zamudio
,
R.
Vega-Sanchez
,
F.
Vadillo-Ortega
.
2009
.
Fetal membranes exhibit selective leukocyte chemotaxic activity during human labor.
J. Reprod. Immunol.
80
:
122
131
.
41.
Gomez-Lopez
N.
,
L.
Vadillo-Perez
,
A.
Hernandez-Carbajal
,
M.
Godines-Enriquez
,
D. M.
Olson
,
F.
Vadillo-Ortega
.
2011
.
Specific inflammatory microenvironments in the zones of the fetal membranes at term delivery.
Am. J. Obstet. Gynecol.
205
:
235.e15
235.e24
.
42.
Gomez-Lopez
N.
,
L.
Vadillo-Perez
,
S.
Nessim
,
D. M.
Olson
,
F.
Vadillo-Ortega
.
2011
.
Choriodecidua and amnion exhibit selective leukocyte chemotaxis during term human labor.
Am. J. Obstet. Gynecol.
204
:
364.e9
364.e16
.
43.
Gomez-Lopez
N.
,
R.
Vega-Sanchez
,
M.
Castillo-Castrejon
,
R.
Romero
,
K.
Cubeiro-Arreola
,
F.
Vadillo-Ortega
.
2013
.
Evidence for a role for the adaptive immune response in human term parturition.
Am. J. Reprod. Immunol.
69
:
212
230
.
44.
Leng
Y.
,
R.
Romero
,
Y.
Xu
,
J.
Galaz
,
R.
Slutsky
,
M.
Arenas-Hernandez
,
V.
Garcia-Flores
,
K.
Motomura
,
S. S.
Hassan
,
A.
Reboldi
,
N.
Gomez-Lopez
.
2019
.
Are B cells altered in the decidua of women with preterm or term labor?
Am. J. Reprod. Immunol.
81
:
e13102
.
45.
Miller
D.
,
M.
Gershater
,
R.
Slutsky
,
R.
Romero
,
N.
Gomez-Lopez
.
2020
.
Maternal and fetal T cells in term pregnancy and preterm labor.
Cell. Mol. Immunol.
17
:
693
704
.
46.
Arenas-Hernandez
M.
,
N.
Gomez-Lopez
,
V.
Garcia-Flores
,
C.
Rangel-Escareño
,
L. M.
Alvarez-Salas
,
N.
Martinez-Acuña
,
J. A.
Vazquez-Perez
,
R.
Vega-Sanchez
.
2019
.
Choriodecidual leukocytes display a unique gene expression signature in spontaneous labor at term.
Genes Immun.
20
:
56
68
.
47.
Gomez-Lopez
N.
,
D.
StLouis
,
M. A.
Lehr
,
E. N.
Sanchez-Rodriguez
,
M.
Arenas-Hernandez
.
2014
.
Immune cells in term and preterm labor.
Cell. Mol. Immunol.
11
:
571
581
.
48.
St. Louis
D.
,
R.
Romero
,
O.
Plazyo
,
M.
Arenas-Hernandez
,
B.
Panaitescu
,
Y.
Xu
,
T.
Milovic
,
Z.
Xu
,
G.
Bhatti
,
Q. S.
Mi
, et al
2016
.
Invariant NKT cell activation induces late preterm birth that is attenuated by rosiglitazone.
J. Immunol.
196
:
1044
1059
.
49.
Xu
Y.
,
R.
Romero
,
D.
Miller
,
L.
Kadam
,
T. N.
Mial
,
O.
Plazyo
,
V.
Garcia-Flores
,
S. S.
Hassan
,
Z.
Xu
,
A. L.
Tarca
, et al
2016
.
An M1-like macrophage polarization in decidual tissue during spontaneous preterm labor that is attenuated by rosiglitazone treatment.
J. Immunol.
196
:
2476
2491
.
50.
Arenas-Hernandez
M.
,
R.
Romero
,
Y.
Xu
,
B.
Panaitescu
,
V.
Garcia-Flores
,
D.
Miller
,
H.
Ahn
,
B.
Done
,
S. S.
Hassan
,
C. D.
Hsu
, et al
2019
.
Effector and activated T cells induce preterm labor and birth that is prevented by treatment with progesterone.
J. Immunol.
202
:
2585
2608
.
51.
Slutsky
R.
,
R.
Romero
,
Y.
Xu
,
J.
Galaz
,
D.
Miller
,
B.
Done
,
A. L.
Tarca
,
S.
Gregor
,
S. S.
Hassan
,
Y.
Leng
,
N.
Gomez-Lopez
.
2019
.
Exhausted and senescent T cells at the maternal-fetal interface in preterm and term labor.
J. Immunol. Res.
2019
:
3128010
.
52.
Tang
F.
,
C.
Barbacioru
,
Y.
Wang
,
E.
Nordman
,
C.
Lee
,
N.
Xu
,
X.
Wang
,
J.
Bodeau
,
B. B.
Tuch
,
A.
Siddiqui
, et al
2009
.
mRNA-Seq whole-transcriptome analysis of a single cell.
Nat. Methods
6
:
377
382
.
53.
Hashimshony
T.
,
F.
Wagner
,
N.
Sher
,
I.
Yanai
.
2012
.
CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification.
Cell Rep.
2
:
666
673
.
54.
Ramsköld
D.
,
S.
Luo
,
Y. C.
Wang
,
R.
Li
,
Q.
Deng
,
O. R.
Faridani
,
G. A.
Daniels
,
I.
Khrebtukova
,
J. F.
Loring
,
L. C.
Laurent
, et al
2012
.
Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells. [Published erratum appears in 2020 Nat. Biotechnol. 38: 374.]
Nat. Biotechnol.
30
:
777
782
.
55.
Jaitin
D. A.
,
E.
Kenigsberg
,
H.
Keren-Shaul
,
N.
Elefant
,
F.
Paul
,
I.
Zaretsky
,
A.
Mildner
,
N.
Cohen
,
S.
Jung
,
A.
Tanay
,
I.
Amit
.
2014
.
Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types.
Science
343
:
776
779
.
56.
Klein
A. M.
,
L.
Mazutis
,
I.
Akartuna
,
N.
Tallapragada
,
A.
Veres
,
V.
Li
,
L.
Peshkin
,
D. A.
Weitz
,
M. W.
Kirschner
.
2015
.
Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells.
Cell
161
:
1187
1201
.
57.
Macosko
E. Z.
,
A.
Basu
,
R.
Satija
,
J.
Nemesh
,
K.
Shekhar
,
M.
Goldman
,
I.
Tirosh
,
A. R.
Bialas
,
N.
Kamitaki
,
E. M.
Martersteck
, et al
2015
.
Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets.
Cell
161
:
1202
1214
.
58.
Mora-Castilla
S.
,
C.
To
,
S.
Vaezeslami
,
R.
Morey
,
S.
Srinivasan
,
J. N.
Dumdie
,
H.
Cook-Andersen
,
J.
Jenkins
,
L. C.
Laurent
.
2016
.
Miniaturization technologies for efficient single-cell library preparation for next-generation sequencing.
J. Lab. Autom.
21
:
557
567
.
59.
Zheng
G. X.
,
J. M.
Terry
,
P.
Belgrader
,
P.
Ryvkin
,
Z. W.
Bent
,
R.
Wilson
,
S. B.
Ziraldo
,
T. D.
Wheeler
,
G. P.
McDermott
,
J.
Zhu
, et al
2017
.
Massively parallel digital transcriptional profiling of single cells.
Nat. Commun.
8
:
14049
.
60.
Aldridge
S.
,
S. A.
Teichmann
.
2020
.
Single cell transcriptomics comes of age.
Nat. Commun.
11
:
4307
.
61.
Vazquez
J.
,
I. M.
Ong
,
A. K.
Stanic
.
2019
.
Single-cell technologies in reproductive immunology.
Am. J. Reprod. Immunol.
82
:
e13157
.
62.
Li
H.
,
Q.
Huang
,
Y.
Liu
,
L. X.
Garmire
.
2020
.
Single cell transcriptome research in human placenta.
Reproduction
160
:
R155
R167
.
63.
Krjutškov
K.
,
S.
Katayama
,
M.
Saare
,
M.
Vera-Rodriguez
,
D.
Lubenets
,
K.
Samuel
,
T.
Laisk-Podar
,
H.
Teder
,
E.
Einarsdottir
,
A.
Salumets
,
J.
Kere
.
2016
.
Single-cell transcriptome analysis of endometrial tissue.
Hum. Reprod.
31
:
844
853
.
64.
Wang
W.
,
F.
Vilella
,
P.
Alama
,
I.
Moreno
,
M.
Mignardi
,
A.
Isakova
,
W.
Pan
,
C.
Simon
,
S. R.
Quake
.
2020
.
Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle.
Nat. Med.
26
:
1644
1653
.
65.
Vilella
F.
,
C.
Simon
.
2021
.
Reproductive medicine, as seen through single-cell glasses.
Fertil. Steril.
115
:
296
297
.
66.
Liu
Y.
,
X.
Fan
,
R.
Wang
,
X.
Lu
,
Y. L.
Dang
,
H.
Wang
,
H. Y.
Lin
,
C.
Zhu
,
H.
Ge
,
J. C.
Cross
,
H.
Wang
.
2018
.
Single-cell RNA-seq reveals the diversity of trophoblast subtypes and patterns of differentiation in the human placenta.
Cell Res.
28
:
819
832
.
67.
Suryawanshi
H.
,
P.
Morozov
,
A.
Straus
,
N.
Sahasrabudhe
,
K. E. A.
Max
,
A.
Garzia
,
M.
Kustagi
,
T.
Tuschl
,
Z.
Williams
.
2018
.
A single-cell survey of the human first-trimester placenta and decidua.
Sci. Adv.
4
:
eaau4788
.
68.
Vento-Tormo
R.
,
M.
Efremova
,
R. A.
Botting
,
M. Y.
Turco
,
M.
Vento-Tormo
,
K. B.
Meyer
,
J. E.
Park
,
E.
Stephenson
,
K.
Polański
,
A.
Goncalves
, et al
2018
.
Single-cell reconstruction of the early maternal-fetal interface in humans.
Nature
563
:
347
353
.
69.
Sun
T.
,
T. L.
Gonzalez
,
N.
Deng
,
R.
DiPentino
,
E. L.
Clark
,
B.
Lee
,
J.
Tang
,
Y.
Wang
,
B. R.
Stripp
,
C.
Yao
, et al
2020
.
Sexually dimorphic crosstalk at the maternal-fetal interface.
J. Clin. Endocrinol. Metab.
105
:
e4831
.
70.
Yi
Y.
,
H.
Zhu
,
C.
Klausen
,
P. C. K.
Leung
.
2021
.
Transcription factor SOX4 facilitates BMP2-regulated gene expression during invasive trophoblast differentiation.
FASEB J.
35
:
e22028
.
71.
Du
L.
,
W.
Deng
,
S.
Zeng
,
P.
Xu
,
L.
Huang
,
Y.
Liang
,
Y.
Wang
,
H.
Xu
,
J.
Tang
,
S.
Bi
, et al
2021
.
Single-cell transcriptome analysis reveals defective decidua stromal niche attributes to recurrent spontaneous abortion.
Cell Prolif.
54
:
e13125
.
72.
He
J. P.
,
Q.
Tian
,
Q. Y.
Zhu
,
J. L.
Liu
.
2021
.
Identification of intercellular crosstalk between decidual cells and niche cells in mice.
Int. J. Mol. Sci.
22
:
7696
.
73.
Wang
F.
,
W.
Jia
,
M.
Fan
,
X.
Shao
,
Z.
Li
,
Y.
Liu
,
Y.
Ma
,
Y. X.
Li
,
R.
Li
,
Q.
Tu
,
Y. L.
Wang
.
2021
.
Single-cell immune landscape of human recurrent miscarriage.
Genomics Proteomics Bioinformatics
19
:
208
222
.
74.
Guo
C.
,
P.
Cai
,
L.
Jin
,
Q.
Sha
,
Q.
Yu
,
W.
Zhang
,
C.
Jiang
,
Q.
Liu
,
D.
Zong
,
K.
Li
, et al
2021
.
Single-cell profiling of the human decidual immune microenvironment in patients with recurrent pregnancy loss.
Cell Discov.
7
:
1
.
75.
Chen
P.
,
L.
Zhou
,
J.
Chen
,
Y.
Lu
,
C.
Cao
,
S.
Lv
,
Z.
Wei
,
L.
Wang
,
J.
Chen
,
X.
Hu
, et al
2021
.
The immune atlas of human deciduas with unexplained recurrent pregnancy loss.
Front. Immunol.
12
:
689019
.
76.
Shannon
M. J.
,
J.
Baltayeva
,
B.
Castellana
,
J.
Wächter
,
G. L.
McNeill
,
J. S.
Yoon
,
J.
Treissman
,
H. T.
Le
,
P. M.
Lavoie
,
A. G.
Beristain
.
2022
.
Cell trajectory modeling identifies a primitive trophoblast state defined by BCAM enrichment.
Development
149
:
dev199840
.
77.
Ma
W.
,
M.
Cao
,
S.
Bi
,
L.
Du
,
J.
Chen
,
H.
Wang
,
Y.
Jiang
,
Y.
Wu
,
Y.
Liao
,
S.
Kong
,
J.
Liu
.
2022
.
MAX deficiency impairs human endometrial decidualization through down-regulating OSR2 in women with recurrent spontaneous abortion.
Cell Tissue Res.
388
:
453
469
.
78.
Pavličev
M.
,
G. P.
Wagner
,
A. R.
Chavan
,
K.
Owens
,
J.
Maziarz
,
C.
Dunn-Fletcher
,
S. G.
Kallapur
,
L.
Muglia
,
H.
Jones
.
2017
.
Single-cell transcriptomics of the human placenta: inferring the cell communication network of the maternal-fetal interface.
Genome Res.
27
:
349
361
.
79.
Tsang
J. C. H.
,
J. S. L.
Vong
,
L.
Ji
,
L. C. Y.
Poon
,
P.
Jiang
,
K. O.
Lui
,
Y. B.
Ni
,
K. F.
To
,
Y. K. Y.
Cheng
,
R. W. K.
Chiu
,
Y. M. D.
Lo
.
2017
.
Integrative single-cell and cell-free plasma RNA transcriptomics elucidates placental cellular dynamics.
Proc. Natl. Acad. Sci. USA
114
:
E7786
E7795
.
80.
Pique-Regi
R.
,
R.
Romero
,
A. L.
Tarca
,
E. D.
Sendler
,
Y.
Xu
,
V.
Garcia-Flores
,
Y.
Leng
,
F.
Luca
,
S. S.
Hassan
,
N.
Gomez-Lopez
.
2019
.
Single cell transcriptional signatures of the human placenta in term and preterm parturition.
eLife
8
:
e52004
.
81.
Liang
G.
,
C.
Zhou
,
X.
Jiang
,
Y.
Zhang
,
B.
Huang
,
S.
Gao
,
Z.
Kang
,
D.
Ma
,
F.
Wang
,
B.
Gottgens
, et al
2021
.
De novo generation of macrophage from placenta-derived hemogenic endothelium.
Dev. Cell
56
:
2121
2133.e6
.
82.
Marsh
B.
,
R.
Blelloch
.
2020
.
Single nuclei RNA-seq of mouse placental labyrinth development.
eLife
9
:
e60266
.
83.
Nelson
A. C.
,
A. W.
Mould
,
E. K.
Bikoff
,
E. J.
Robertson
.
2016
.
Single-cell RNA-seq reveals cell type-specific transcriptional signatures at the maternal-foetal interface during pregnancy. [Published erratum appears in 2018 Nat. Commun. 9: 16219.]
Nat. Commun.
7
:
11414
.
84.
Toothaker
J. M.
,
O.
Olaloye
,
B. T.
McCourt
,
C. C.
McCourt
,
T. N.
Silva
,
R. M.
Case
,
P.
Liu
,
D.
Yimlamai
,
G.
Tseng
,
L.
Konnikova
.
2022
.
Immune landscape of human placental villi using single-cell analysis.
Development
149
:
dev200013
.
85.
Zhou
X.
,
Y.
Xu
,
S.
Ren
,
D.
Liu
,
N.
Yang
,
Q.
Han
,
S.
Kong
,
H.
Wang
,
W.
Deng
,
H.
Qi
,
J.
Lu
.
2021
.
Single-cell RNA-seq revealed diverse cell types in the mouse placenta at mid-gestation.
Exp. Cell Res.
405
:
112715
.
86.
Huang
J. R.
,
J.
Nie
,
L. J.
Liu
,
X. W.
Zhang
,
Y. M.
Xie
,
Q. Z.
Peng
,
W. N.
Wang
,
C. L.
Pei
,
Y. H.
Zhao
,
R.
Liu
, et al
2020
.
Single-cell transcriptomics reveals the heterogeneity of the decidual endothelial cells that participate in labor onset.
Eur. Rev. Med. Pharmacol. Sci.
24
:
10359
10365
.
87.
Huang
J.
,
Q.
Li
,
Q.
Peng
,
Y.
Xie
,
W.
Wang
,
C.
Pei
,
Y.
Zhao
,
R.
Liu
,
L.
Huang
,
T.
Li
, et al
2021
.
Single-cell RNA sequencing reveals heterogeneity and differential expression of decidual tissues during the peripartum period.
Cell Prolif.
54
:
e12967
.
88.
Huang
J.
,
W.
Zhang
,
Y.
Zhao
,
J.
Li
,
M.
Xie
,
Y.
Lu
,
Q.
Peng
,
J.
Zhang
,
P.
Li
,
L.
Dai
.
2021
.
Deciphering the intercellular communication network of peripartum decidua that orchestrates delivery.
Front. Cell Dev. Biol.
9
:
770621
.
89.
Pique-Regi
R.
,
R.
Romero
,
V.
Garcia-Flores
,
A.
Peyvandipour
,
A. L.
Tarca
,
E.
Pusod
,
J.
Galaz
,
D.
Miller
,
G.
Bhatti
,
R.
Para
, et al
2022
.
A single-cell atlas of the myometrium in human parturition.
JCI Insight
7
:
e153921
.
90.
Garcia-Flores
V.
,
R.
Romero
,
A.
Peyvandipour
,
J.
Galaz
,
E.
Pusod
,
B.
Panaitescu
,
D.
Miller
,
Y.
Xu
,
L.
Tao
,
Y.
Liu
, et al
2022
.
The single-cell atlas of the murine reproductive tissues during preterm labor.
bioRxiv
:
2022.2004.2027.489704
.
91.
Pique-Regi
R.
,
R.
Romero
,
A. L.
Tarca
,
F.
Luca
,
Y.
Xu
,
A.
Alazizi
,
Y.
Leng
,
C. D.
Hsu
,
N.
Gomez-Lopez
.
2020
.
Does the human placenta express the canonical cell entry mediators for SARS-CoV-2?
eLife
9
:
e58716
.
92.
Li
M.
,
L.
Chen
,
J.
Zhang
,
C.
Xiong
,
X.
Li
.
2020
.
The SARS-CoV-2 receptor ACE2 expression of maternal-fetal interface and fetal organs by single-cell transcriptome study.
PLoS One
15
:
e0230295
.
93.
Lu-Culligan
A.
,
A. R.
Chavan
,
P.
Vijayakumar
,
L.
Irshaid
,
E. M.
Courchaine
,
K. M.
Milano
,
Z.
Tang
,
S. D.
Pope
,
E.
Song
,
C. B. F.
Vogels
, et al
Yale IMPACT Team
.
2021
.
Maternal respiratory SARS-CoV-2 infection in pregnancy is associated with a robust inflammatory response at the maternal-fetal interface.
Med (N Y)
2
:
591
610.e10
.
94.
Constantino
F. B.
,
S. S.
Cury
,
C. R.
Nogueira
,
R. F.
Carvalho
,
L. A.
Justulin
.
2021
.
Prediction of non-canonical routes for SARS-CoV-2 infection in human placenta cells.
Front. Mol. Biosci.
8
:
614728
.
95.
Cui
D.
,
Y.
Liu
,
X.
Jiang
,
C.
Ding
,
L. C.
Poon
,
H.
Wang
,
H.
Yang
.
2021
.
Single-cell RNA expression profiling of SARS-CoV-2-related ACE2 and TMPRSS2 in human trophectoderm and placenta.
Ultrasound Obstet. Gynecol.
57
:
248
256
.
96.
Li
Q.
,
W.
Wang
,
C.
Pei
,
Y.
Zhao
,
R.
Liu
,
W.
Zhang
,
L.
Huang
,
T.
Li
,
J.
Huang
.
2021
.
Expression of SARS-CoV-2 entry genes ACE2 and TMPRSS2 at single cell resolution in the peripartum decidua.
Am. J. Transl. Res.
13
:
4389
4400
.
97.
Garcia-Flores
V.
,
R.
Romero
,
Y.
Xu
,
K. R.
Theis
,
M.
Arenas-Hernandez
,
D.
Miller
,
A.
Peyvandipour
,
G.
Bhatti
,
J.
Galaz
,
M.
Gershater
, et al
2022
.
Maternal-fetal immune responses in pregnant women infected with SARS-CoV-2.
Nat. Commun.
13
:
320
.
98.
Chen
J.
,
L.
Du
,
F.
Wang
,
X.
Shao
,
X.
Wang
,
W.
Yu
,
S.
Bi
,
D.
Chen
,
X.
Pan
,
S.
Zeng
, et al
2022
.
Cellular and molecular atlas of the placenta from a COVID-19 pregnant woman infected at midgestation highlights the defective impacts on foetal health.
Cell Prolif.
55
:
e13204
.
99.
Huang
Z.
,
S.
Xia
,
S.
Mei
,
Y.
Wen
,
J.
Liu
,
C.
Dong
,
W.
Chen
,
P.
Yu
,
L.
Qu
,
Y.
Luo
,
L.
Zheng
.
2022
.
Integrated analysis reveals the characteristics and effects of SARS-CoV-2 maternal-fetal transmission.
Front. Microbiol.
13
:
813187
.
100.
Pijnenborg
R.
,
L.
Vercruysse
,
I.
Brosens
.
2011
.
Deep placentation.
Best Pract. Res. Clin. Obstet. Gynaecol.
25
:
273
285
.
101.
Burton
G. J.
,
E.
Jauniaux
.
2015
.
What is the placenta?
Am. J. Obstet. Gynecol.
213
(
4 Suppl
):
S6.e1
S6.e4
.
102.
Brosens
I.
,
P.
Puttemans
,
G.
Benagiano
.
2019
.
Placental bed research: I. The placental bed: from spiral arteries remodeling to the great obstetrical syndromes.
Am. J. Obstet. Gynecol.
221
:
437
456
.
103.
Albrecht
E. D.
,
G. J.
Pepe
.
2020
.
Regulation of uterine spiral artery remodeling: a review.
Reprod. Sci.
27
:
1932
1942
.
104.
Sato
Y.
2020
.
Endovascular trophoblast and spiral artery remodeling.
Mol. Cell. Endocrinol.
503
:
110699
.
105.
Lu
X.
,
R.
Wang
,
C.
Zhu
,
H.
Wang
,
H. Y.
Lin
,
Y.
Gu
,
J. C.
Cross
,
H.
Wang
.
2017
.
Fine-tuned and cell-cycle-restricted expression of fusogenic protein syncytin-2 maintains functional placental syncytia. [Published erratum appears in 2018 Cell Rep. 23: 3979.]
Cell Rep.
21
:
1150
1159
.
106.
Haider
S.
,
G.
Meinhardt
,
L.
Saleh
,
C.
Fiala
,
J.
Pollheimer
,
M.
Knöfler
.
2016
.
Notch1 controls development of the extravillous trophoblast lineage in the human placenta.
Proc. Natl. Acad. Sci. USA
113
:
E7710
E7719
.
107.
Saelens
W.
,
R.
Cannoodt
,
H.
Todorov
,
Y.
Saeys
.
2019
.
A comparison of single-cell trajectory inference methods.
Nat. Biotechnol.
37
:
547
554
.
108.
Henry
V. J.
,
A. E.
Bandrowski
,
A. S.
Pepin
,
B. J.
Gonzalez
,
A.
Desfeux
.
2014
.
OMICtools: an informative directory for multi-omic data analysis.
Database (Oxford)
2014
:
bau069
.
109.
Zappia
L.
,
B.
Phipson
,
A.
Oshlack
.
2018
.
Exploring the single-cell RNA-seq analysis landscape with the scRNA-tools database.
PLoS Comput. Biol.
14
:
e1006245
.
110.
Farley
A. E.
,
C. H.
Graham
,
G. N.
Smith
.
2004
.
Contractile properties of human placental anchoring villi.
Am. J. Physiol. Regul. Integr. Comp. Physiol.
287
:
R680
R685
.
111.
Thornburg
K. L.
,
N.
Marshall
.
2015
.
The placenta is the center of the chronic disease universe.
Am. J. Obstet. Gynecol.
213
(
4 Suppl
):
S14
S20
.
112.
Krop
J.
,
A.
van der Zwan
,
M. E.
Ijsselsteijn
,
H.
Kapsenberg
,
S. J.
Luk
,
S. H.
Hendriks
,
C.
van der Keur
,
L. J.
Verleng
,
A.
Somarakis
,
L.
van der Meeren
, et al
2022
.
Imaging mass cytometry reveals the prominent role of myeloid cells at the maternal-fetal interface.
iScience
25
:
104648
.
113.
Li
R.
,
T. Y.
Wang
,
X.
Xu
,
O.
Emery
,
M.
Yi
,
S. P.
Wu
,
F. J.
DeMayo
.
2022
.
Spatial transcriptomic profiles of mouse uterine microenvironments at pregnancy day 7.5.
Biol. Reprod.
107
:
529
545
.
114.
Wang
Y.
,
R.
Wang
,
S.
Zhang
,
S.
Song
,
C.
Jiang
,
G.
Han
,
M.
Wang
,
J.
Ajani
,
A.
Futreal
,
L.
Wang
.
2019
.
iTALK: an R package to characterize and illustrate intercellular communication.
bioRxiv
:
507871
.
115.
Browaeys
R.
,
W.
Saelens
,
Y.
Saeys
.
2020
.
NicheNet: modeling intercellular communication by linking ligands to target genes.
Nat. Methods
17
:
159
162
.
116.
Cabello-Aguilar
S.
,
M.
Alame
,
F.
Kon-Sun-Tack
,
C.
Fau
,
M.
Lacroix
,
J.
Colinge
.
2020
.
SingleCellSignalR: inference of intercellular networks from single-cell transcriptomics.
Nucleic Acids Res.
48
:
e55
.
117.
Efremova
M.
,
M.
Vento-Tormo
,
S. A.
Teichmann
,
R.
Vento-Tormo
.
2020
.
CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes.
Nat. Protoc.
15
:
1484
1506
.
118.
Jin
S.
,
C. F.
Guerrero-Juarez
,
L.
Zhang
,
I.
Chang
,
R.
Ramos
,
C. H.
Kuan
,
P.
Myung
,
M. V.
Plikus
,
Q.
Nie
.
2021
.
Inference and analysis of cell-cell communication using CellChat.
Nat. Commun.
12
:
1088
.
119.
Hiby
S. E.
,
R.
Apps
,
A. M.
Sharkey
,
L. E.
Farrell
,
L.
Gardner
,
A.
Mulder
,
F. H.
Claas
,
J. J.
Walker
,
C. W.
Redman
,
L.
Morgan
, et al
2010
.
Maternal activating KIRs protect against human reproductive failure mediated by fetal HLA-C2. [Published erratum appears in 2011 J. Clin. Invest. 121: 455.]
J. Clin. Invest.
120
:
4102
4110
.
120.
Colucci
F.
2017
.
The role of KIR and HLA interactions in pregnancy complications.
Immunogenetics
69
:
557
565
.
121.
Huhn
O.
,
O.
Chazara
,
M. A.
Ivarsson
,
C.
Retière
,
T. C.
Venkatesan
,
P. J.
Norman
,
H. G.
Hilton
,
J.
Jayaraman
,
J. A.
Traherne
,
J.
Trowsdale
, et al
2018
.
High-resolution genetic and phenotypic analysis of KIR2DL1 alleles and their association with pre-eclampsia.
J. Immunol.
201
:
2593
2601
.
122.
Johnsen
G. M.
,
G. L.
Størvold
,
J. J. M.
Drabbels
,
G. W.
Haasnoot
,
M.
Eikmans
,
M. J.
Spruyt-Gerritse
,
P.
Alnæs-Katjavivi
,
S. A.
Scherjon
,
C. W. G.
Redman
,
F. H. J.
Claas
,
A. C.
Staff
.
2018
.
The combination of maternal KIR-B and fetal HLA-C2 is associated with decidua basalis acute atherosis in pregnancies with preeclampsia.
J. Reprod. Immunol.
129
:
23
29
.
123.
Repnik
U.
,
T.
Tilburgs
,
D. L.
Roelen
,
B. J.
van der Mast
,
H. H.
Kanhai
,
S.
Scherjon
,
F. H.
Claas
.
2008
.
Comparison of macrophage phenotype between decidua basalis and decidua parietalis by flow cytometry.
Placenta
29
:
405
412
.
124.
Kwan
M.
,
A.
Hazan
,
J.
Zhang
,
R. L.
Jones
,
L. K.
Harris
,
W.
Whittle
,
S.
Keating
,
C. E.
Dunk
,
S. J.
Lye
.
2014
.
Dynamic changes in maternal decidual leukocyte populations from first to second trimester gestation.
Placenta
35
:
1027
1034
.
125.
Shah
N. M.
,
A. A.
Herasimtschuk
,
A.
Boasso
,
A.
Benlahrech
,
D.
Fuchs
,
N.
Imami
,
M. R.
Johnson
.
2017
.
Changes in T cell and dendritic cell phenotype from mid to late pregnancy are indicative of a shift from immune tolerance to immune activation.
Front. Immunol.
8
:
1138
.
126.
van der Zwan
A.
,
V.
van Unen
,
G.
Beyrend
,
S.
Laban
,
C.
van der Keur
,
H. J. M.
Kapsenberg
,
T.
Höllt
,
S. M.
Chuva de Sousa Lopes
,
M. P.
van der Hoorn
,
F.
Koning
, et al
2020
.
Visualizing dynamic changes at the maternal-fetal interface throughout human pregnancy by mass cytometry.
Front. Immunol.
11
:
571300
.
127.
Marsh
B.
,
Y.
Zhou
,
M.
Kapidzic
,
S.
Fisher
,
R.
Blelloch
.
2022
.
Regionally distinct trophoblast regulate barrier function and invasion in the human placenta.
eLife
11
:
e78829
.
128.
Cunningham
F. G.
,
K. J.
Leveno
,
J. S.
Dashe
,
B. L.
Hoffman
,
C. Y.
Spong
,
B. M.
Casey
.
2022
.
Implantation and placental development.
In
Williams Obstetrics
, 26th Ed.
McGraw Hill
,
New York, NY
.
129.
Gomez-Lopez
N.
,
R.
Romero
,
S. S.
Hassan
,
G.
Bhatti
,
S. M.
Berry
,
J. P.
Kusanovic
,
P.
Pacora
,
A. L.
Tarca
.
2019
.
The cellular transcriptome in the maternal circulation during normal pregnancy: a longitudinal study.
Front. Immunol.
10
:
2863
.
130.
Tarca
A. L.
,
R.
Romero
,
Z.
Xu
,
N.
Gomez-Lopez
,
O.
Erez
,
C. D.
Hsu
,
S. S.
Hassan
,
V. J.
Carey
.
2019
.
Targeted expression profiling by RNA-Seq improves detection of cellular dynamics during pregnancy and identifies a role for T cells in term parturition.
Sci. Rep.
9
:
848
.
131.
Tarca
A. L.
,
R.
Romero
,
R.
Pique-Regi
,
P.
Pacora
,
B.
Done
,
M.
Kacerovsky
,
G.
Bhatti
,
S.
Jaiman
,
S. S.
Hassan
,
C. D.
Hsu
,
N.
Gomez-Lopez
.
2020
.
Amniotic fluid cell-free transcriptome: a glimpse into fetal development and placental cellular dynamics during normal pregnancy.
BMC Med. Genomics
13
:
25
.
132.
Lo
Y. M.
,
N.
Corbetta
,
P. F.
Chamberlain
,
V.
Rai
,
I. L.
Sargent
,
C. W.
Redman
,
J. S.
Wainscoat
.
1997
.
Presence of fetal DNA in maternal plasma and serum.
Lancet
350
:
485
487
.
133.
Lo
Y. M.
,
T. K.
Lau
,
J.
Zhang
,
T. N.
Leung
,
A. M.
Chang
,
N. M.
Hjelm
,
R. S.
Elmes
,
D. W.
Bianchi
.
1999
.
Increased fetal DNA concentrations in the plasma of pregnant women carrying fetuses with trisomy 21.
Clin. Chem.
45
:
1747
1751
.
134.
Ariga
H.
,
H.
Ohto
,
M. P.
Busch
,
S.
Imamura
,
R.
Watson
,
W.
Reed
,
T. H.
Lee
.
2001
.
Kinetics of fetal cellular and cell-free DNA in the maternal circulation during and after pregnancy: implications for noninvasive prenatal diagnosis.
Transfusion
41
:
1524
1530
.
135.
Bianchi
D. W.
,
Y. M.
Lo
.
2001
.
Fetomaternal cellular and plasma DNA trafficking: the Yin and the Yang.
Ann. N. Y. Acad. Sci.
945
:
119
131
.
136.
Ng
E. K.
,
N. B.
Tsui
,
T. K.
Lau
,
T. N.
Leung
,
R. W.
Chiu
,
N. S.
Panesar
,
L. C.
Lit
,
K. W.
Chan
,
Y. M.
Lo
.
2003
.
mRNA of placental origin is readily detectable in maternal plasma.
Proc. Natl. Acad. Sci. USA
100
:
4748
4753
.
137.
Chiu
R. W.
,
W. B.
Lui
,
M. C.
Cheung
,
N.
Kumta
,
A.
Farina
,
I.
Banzola
,
S.
Grotti
,
N.
Rizzo
,
C. J.
Haines
,
Y. M.
Lo
.
2006
.
Time profile of appearance and disappearance of circulating placenta-derived mRNA in maternal plasma.
Clin. Chem.
52
:
313
316
.
138.
Taglauer
E. S.
,
L.
Wilkins-Haug
,
D. W.
Bianchi
.
2014
.
Review: cell-free fetal DNA in the maternal circulation as an indication of placental health and disease.
Placenta
35
(
Suppl
):
S64
S68
.
139.
Yeganeh Kazemi
N.
,
B.
Fedyshyn
,
S.
Sutor
,
Y.
Fedyshyn
,
S.
Markovic
,
E. A. L.
Enninga
.
2021
.
Maternal monocytes respond to cell-free fetal DNA and initiate key processes of human parturition.
J. Immunol.
207
:
2433
2444
.
140.
Gomez-Lopez
N.
,
L. J.
Guilbert
,
D. M.
Olson
.
2010
.
Invasion of the leukocytes into the fetal-maternal interface during pregnancy.
J. Leukoc. Biol.
88
:
625
633
.
141.
Li
J.
,
Q.
Wang
,
Y.
An
,
X.
Chen
,
Y.
Xing
,
Q.
Deng
,
Z.
Li
,
S.
Wang
,
X.
Dai
,
N.
Liang
, et al
2022
.
Integrative single-cell RNA-Seq and ATAC-Seq analysis of mesenchymal stem/stromal cells derived from human placenta.
Front. Cell Dev. Biol.
10
:
836887
.
142.
Miller
D.
,
K.
Motomura
,
V.
Garcia-Flores
,
R.
Romero
,
N.
Gomez-Lopez
.
2018
.
Innate lymphoid cells in the maternal and fetal compartments.
Front. Immunol.
9
:
2396
.
143.
Osman
I.
,
A.
Young
,
M. A.
Ledingham
,
A. J.
Thomson
,
F.
Jordan
,
I. A.
Greer
,
J. E.
Norman
.
2003
.
Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term.
Mol. Hum. Reprod.
9
:
41
45
.
144.
Hamilton
S.
,
Y.
Oomomian
,
G.
Stephen
,
O.
Shynlova
,
C. L.
Tower
,
A.
Garrod
,
S. J.
Lye
,
R. L.
Jones
.
2012
.
Macrophages infiltrate the human and rat decidua during term and preterm labor: evidence that decidual inflammation precedes labor.
Biol. Reprod.
86
:
39
.
145.
Hamilton
S. A.
,
C. L.
Tower
,
R. L.
Jones
.
2013
.
Identification of chemokines associated with the recruitment of decidual leukocytes in human labour: potential novel targets for preterm labour.
PLoS One
8
:
e56946
.
146.
Shynlova
O.
,
T.
Nedd-Roderique
,
Y.
Li
,
A.
Dorogin
,
T.
Nguyen
,
S. J.
Lye
.
2013
.
Infiltration of myeloid cells into decidua is a critical early event in the labour cascade and post-partum uterine remodelling.
J. Cell. Mol. Med.
17
:
311
324
.
147.
Provine
N. M.
,
P.
Klenerman
.
2020
.
MAIT cells in health and disease.
Annu. Rev. Immunol.
38
:
203
228
.
148.
Solders
M.
,
L.
Gorchs
,
T.
Erkers
,
A. C.
Lundell
,
S.
Nava
,
S.
Gidlöf
,
E.
Tiblad
,
I.
Magalhaes
,
H.
Kaipe
.
2017
.
MAIT cells accumulate in placental intervillous space and display a highly cytotoxic phenotype upon bacterial stimulation.
Sci. Rep.
7
:
6123
.
149.
Solders
M.
,
L.
Gorchs
,
S.
Gidlöf
,
E.
Tiblad
,
A. C.
Lundell
,
H.
Kaipe
.
2017
.
Maternal adaptive immune cells in decidua parietalis display a more activated and coinhibitory phenotype compared to decidua basalis.
Stem Cells Int.
2017
:
8010961
.
150.
Solders
M.
,
L.
Gorchs
,
E.
Tiblad
,
S.
Gidlöf
,
E.
Leeansyah
,
J.
Dias
,
J. K.
Sandberg
,
I.
Magalhaes
,
A. C.
Lundell
,
H.
Kaipe
.
2019
.
Recruitment of MAIT cells to the intervillous space of the placenta by placenta-derived chemokines.
Front. Immunol.
10
:
1300
.
151.
Vazquez
J.
,
M.
Chavarria
,
D. A.
Chasman
,
R.
Welch Schwartz
,
C. T.
Tyler
,
G.
Lopez
,
R. C.
Fisher
,
I. M.
Ong
,
A. K.
Stanic
.
2021
.
Multiomic analysis reveals decidual-specific transcriptional programing of MAIT cells.
Am. J. Reprod. Immunol.
86
:
e13495
.
152.
Romero
R.
,
S. K.
Dey
,
S. J.
Fisher
.
2014
.
Preterm labor: one syndrome, many causes.
Science
345
:
760
765
.
153.
Jung
E.
,
R.
Romero
,
L.
Yeo
,
N.
Gomez-Lopez
,
P.
Chaemsaithong
,
A.
Jaovisidha
,
F.
Gotsch
,
O.
Erez
.
2022
.
The etiology of preeclampsia.
Am. J. Obstet. Gynecol.
226
(
2 Suppl
):
S844
S866
.
154.
Zhang
T.
,
Q.
Bian
,
Y.
Chen
,
X.
Wang
,
S.
Yu
,
S.
Liu
,
P.
Ji
,
L.
Li
,
M.
Shrestha
,
S.
Dong
, et al
2021
.
Dissecting human trophoblast cell transcriptional heterogeneity in preeclampsia using single-cell RNA sequencing.
Mol. Genet. Genomic Med.
9
:
e1730
.
155.
Guo
F.
,
B.
Zhang
,
H.
Yang
,
Y.
Fu
,
Y.
Wang
,
J.
Huang
,
M.
Cheng
,
X.
Li
,
Z.
Shen
,
L.
Li
, et al
2021
.
Systemic transcriptome comparison between early- and late-onset pre-eclampsia shows distinct pathology and novel biomarkers.
Cell Prolif.
54
:
e12968
.
156.
Zhou
W.
,
H.
Wang
,
Y.
Yang
,
F.
Guo
,
B.
Yu
,
Z.
Su
.
2022
.
Trophoblast cell subtypes and dysfunction in the placenta of individuals with preeclampsia revealed by single-cell RNA sequencing.
Mol. Cells
45
:
317
328
.
157.
DiFederico
E.
,
O.
Genbacev
,
S. J.
Fisher
.
1999
.
Preeclampsia is associated with widespread apoptosis of placental cytotrophoblasts within the uterine wall.
Am. J. Pathol.
155
:
293
301
.
158.
Leung
D. N.
,
S. C.
Smith
,
K. F.
To
,
D. S.
Sahota
,
P. N.
Baker
.
2001
.
Increased placental apoptosis in pregnancies complicated by preeclampsia.
Am. J. Obstet. Gynecol.
184
:
1249
1250
.
159.
Ishihara
N.
,
H.
Matsuo
,
H.
Murakoshi
,
J. B.
Laoag-Fernandez
,
T.
Samoto
,
T.
Maruo
.
2002
.
Increased apoptosis in the syncytiotrophoblast in human term placentas complicated by either preeclampsia or intrauterine growth retardation.
Am. J. Obstet. Gynecol.
186
:
158
166
.
160.
Kadyrov
M.
,
J. C.
Kingdom
,
B.
Huppertz
.
2006
.
Divergent trophoblast invasion and apoptosis in placental bed spiral arteries from pregnancies complicated by maternal anemia and early-onset preeclampsia/intrauterine growth restriction.
Am. J. Obstet. Gynecol.
194
:
557
563
.
161.
Longtine
M. S.
,
B.
Chen
,
A. O.
Odibo
,
Y.
Zhong
,
D. M.
Nelson
.
2012
.
Villous trophoblast apoptosis is elevated and restricted to cytotrophoblasts in pregnancies complicated by preeclampsia, IUGR, or preeclampsia with IUGR.
Placenta
33
:
352
359
.
162.
von Dadelszen
P.
,
L. A.
Magee
,
J. M.
Roberts
.
2003
.
Subclassification of preeclampsia.
Hypertens. Pregnancy
22
:
143
148
.
163.
Tranquilli
A. L.
,
M. A.
Brown
,
G. G.
Zeeman
,
G.
Dekker
,
B. M.
Sibai
;
Statements from the International Society for the Study of Hypertension in Pregnancy (ISSHP)
.
2013
.
The definition of severe and early-onset preeclampsia.
Pregnancy Hypertens.
3
:
44
47
.
164.
Miller
D.
,
K.
Motomura
,
J.
Galaz
,
M.
Gershater
,
E. D.
Lee
,
R.
Romero
,
N.
Gomez-Lopez
.
2022
.
Cellular immune responses in the pathophysiology of preeclampsia.
J. Leukoc. Biol.
111
:
237
260
.
165.
Chaiworapongsa
T.
,
P.
Chaemsaithong
,
L.
Yeo
,
R.
Romero
.
2014
.
Pre-eclampsia part 1: current understanding of its pathophysiology.
Nat. Rev. Nephrol.
10
:
466
480
.
166.
Burton
G. J.
,
C. W.
Redman
,
J. M.
Roberts
,
A.
Moffett
.
2019
.
Pre-eclampsia: pathophysiology and clinical implications.
BMJ
366
:
l2381
.
167.
Staff
A. C.
,
H. E.
Fjeldstad
,
I. K.
Fosheim
,
K.
Moe
,
G.
Turowski
,
G. M.
Johnsen
,
P.
Alnaes-Katjavivi
,
M.
Sugulle
.
2022
.
Failure of physiological transformation and spiral artery atherosis: their roles in preeclampsia.
Am. J. Obstet. Gynecol.
226
(
2 Suppl
):
S895
S906
.
168.
Norwitz
E. R.
,
J. N.
Robinson
,
J. R.
Challis
.
1999
.
The control of labor.
N. Engl. J. Med.
341
:
660
666
.
169.
Romero
R.
,
J.
Espinoza
,
J. P.
Kusanovic
,
F.
Gotsch
,
S.
Hassan
,
O.
Erez
,
T.
Chaiworapongsa
,
M.
Mazor
.
2006
.
The preterm parturition syndrome.
BJOG
113
(
Suppl 3
):
17
42
.
170.
Smith
R.
2007
.
Parturition.
N. Engl. J. Med.
356
:
271
283
.
171.
Koh
W.
,
A.
Wu
,
L.
Penland
,
B.
Treutlein
,
N. F.
Neff
,
G. L.
Mantalas
,
Y. J.
Blumenfeld
,
Y. Y.
El-Sayed
,
D. K.
Stevenson
,
G. M.
Shaw
,
S. R.
Quake
.
2019
.
Single cell transcriptomes derived from human cervical and uterine tissue during pregnancy.
Adv. Biosyst.
3
:
1800336
.
172.
Blum
J. S.
,
P. A.
Wearsch
,
P.
Cresswell
.
2013
.
Pathways of antigen processing.
Annu. Rev. Immunol.
31
:
443
473
.
173.
Farr
L.
,
S.
Ghosh
,
N.
Jiang
,
K.
Watanabe
,
M.
Parlak
,
R.
Bucala
,
S.
Moonah
.
2020
.
CD74 signaling links inflammation to intestinal epithelial cell regeneration and promotes mucosal healing.
Cell. Mol. Gastroenterol. Hepatol.
10
:
101
112
.
174.
Rodor
J.
,
S. H.
Chen
,
J. P.
Scanlon
,
J. P.
Monteiro
,
A.
Caudrillier
,
S.
Sweta
,
K. R.
Stewart
,
A.
Shmakova
,
R.
Dobie
,
B. E. P.
Henderson
, et al
2021
.
Single-cell RNA-seq profiling of mouse endothelial cells in response to pulmonary arterial hypertension.
Cardiovasc. Res. cvab296.
DOI: 10.1093/cvr/cvab296
.
175.
Romero
R.
,
D. T.
Brody
,
E.
Oyarzun
,
M.
Mazor
,
Y. K.
Wu
,
J. C.
Hobbins
,
S. K.
Durum
.
1989
.
Infection and labor. III. Interleukin-1: a signal for the onset of parturition.
Am. J. Obstet. Gynecol.
160
:
1117
1123
.
176.
Romero
R.
,
S.
Durum
,
C. A.
Dinarello
,
E.
Oyarzun
,
J. C.
Hobbins
,
M. D.
Mitchell
.
1989
.
Interleukin-1 stimulates prostaglandin biosynthesis by human amnion.
Prostaglandins
37
:
13
22
.
177.
Bry
K.
,
M.
Hallman
.
1991
.
Synergistic stimulation of amnion cell prostaglandin E2 synthesis by interleukin-1, tumor necrosis factor and products from activated human granulocytes.
Prostaglandins Leukot. Essent. Fatty Acids
44
:
241
245
.
178.
Romero
R.
,
M.
Mazor
,
F.
Brandt
,
W.
Sepulveda
,
C.
Avila
,
D. B.
Cotton
,
C. A.
Dinarello
.
1992
.
Interleukin-1 alpha and interleukin-1 beta in preterm and term human parturition.
Am. J. Reprod. Immunol.
27
:
117
123
.
179.
Wienke
J.
,
L.
Brouwers
,
L. M.
van der Burg
,
M.
Mokry
,
R. C.
Scholman
,
P. G.
Nikkels
,
B. B.
van Rijn
,
F.
van Wijk
.
2020
.
Human Tregs at the materno-fetal interface show site-specific adaptation reminiscent of tumor Tregs.
JCI Insight
5
:
e137926
.
180.
Shang
J.
,
G.
Ye
,
K.
Shi
,
Y.
Wan
,
C.
Luo
,
H.
Aihara
,
Q.
Geng
,
A.
Auerbach
,
F.
Li
.
2020
.
Structural basis of receptor recognition by SARS-CoV-2.
Nature
581
:
221
224
.
181.
Wang
Q.
,
Y.
Zhang
,
L.
Wu
,
S.
Niu
,
C.
Song
,
Z.
Zhang
,
G.
Lu
,
C.
Qiao
,
Y.
Hu
,
K. Y.
Yuen
, et al
2020
.
Structural and functional basis of SARS-CoV-2 entry by using human ACE2.
Cell
181
:
894
904.e9
.
182.
Hoffmann
M.
,
H.
Kleine-Weber
,
S.
Schroeder
,
N.
Krüger
,
T.
Herrler
,
S.
Erichsen
,
T. S.
Schiergens
,
G.
Herrler
,
N. H.
Wu
,
A.
Nitsche
, et al
2020
.
SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor.
Cell
181
:
271
280.e8
.
183.
Zheng
Q. L.
,
T.
Duan
,
L. P.
Jin
.
2020
.
Single-cell RNA expression profiling of ACE2 and AXL in the human maternal-Fetal interface.
Reprod Dev Med
4
:
7
10
.
184.
Ashary
N.
,
A.
Bhide
,
P.
Chakraborty
,
S.
Colaco
,
A.
Mishra
,
K.
Chhabria
,
M. K.
Jolly
,
D.
Modi
.
2020
.
Single-cell RNA-seq identifies cell subsets in human placenta that highly expresses factors driving pathogenesis of SARS-CoV-2.
Front. Cell Dev. Biol.
8
:
783
.
185.
Beesley
M. A.
,
J. R.
Davidson
,
F.
Panariello
,
S.
Shibuya
,
D.
Scaglioni
,
B. C.
Jones
,
K.
Maksym
,
O.
Ogunbiyi
,
N. J.
Sebire
,
D.
Cacchiarelli
, et al
2022
.
COVID-19 and vertical transmission: assessing the expression of ACE2/TMPRSS2 in the human fetus and placenta to assess the risk of SARS-CoV-2 infection.
BJOG
129
:
256
266
.
186.
See
P.
,
J.
Lum
,
J.
Chen
,
F.
Ginhoux
.
2018
.
A single-cell sequencing guide for immunologists. [Published erratum appears in 2019 Front. Immunol. 10: 278.]
Front. Immunol.
9
:
2425
.
187.
Chen
G.
,
B.
Ning
,
T.
Shi
.
2019
.
Single-cell RNA-Seq technologies and related computational data analysis.
Front. Genet.
10
:
317
.
188.
Lähnemann
D.
,
J.
Köster
,
E.
Szczurek
,
D. J.
McCarthy
,
S. C.
Hicks
,
M. D.
Robinson
,
C. A.
Vallejos
,
K. R.
Campbell
,
N.
Beerenwinkel
,
A.
Mahfouz
, et al
2020
.
Eleven grand challenges in single-cell data science.
Genome Biol.
21
:
31
.
189.
Kharchenko
P. V.
2021
.
The triumphs and limitations of computational methods for scRNA-seq. [Published erratum appears in 2021 Nat. Methods 18: 835.]
Nat. Methods
18
:
723
732
.
190.
Street
K.
,
D.
Risso
,
R. B.
Fletcher
,
D.
Das
,
J.
Ngai
,
N.
Yosef
,
E.
Purdom
,
S.
Dudoit
.
2018
.
Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics.
BMC Genomics
19
:
477
.
191.
Garcia-Flores
V.
,
Y.
Xu
,
E.
Pusod
,
R.
Romero
,
R.
Pique-Regi
,
N.
Gomez-Lopez
.
2022
.
Preparation of single-cell suspensions from the human placenta.
Nat. Protoc.
In press
.
192.
Hafemeister
C.
,
R.
Satija
.
2019
.
Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression.
Genome Biol.
20
:
296
.
193.
Stuart
T.
,
A.
Butler
,
P.
Hoffman
,
C.
Hafemeister
,
E.
Papalexi
,
W. M.
Mauck
3rd
,
Y.
Hao
,
M.
Stoeckius
,
P.
Smibert
,
R.
Satija
.
2019
.
Comprehensive integration of single-cell data.
Cell
177
:
1888
1902.e21
.
194.
Kang
H. M.
,
M.
Subramaniam
,
S.
Targ
,
M.
Nguyen
,
L.
Maliskova
,
E.
McCarthy
,
E.
Wan
,
S.
Wong
,
L.
Byrnes
,
C. M.
Lanata
, et al
2018
.
Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. [Published erratum appears in 2020 Nat. Biotechnol. 38: 1356.]
Nat. Biotechnol.
36
:
89
94
.
195.
Love
M. I.
,
W.
Huber
,
S.
Anders
.
2014
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.
Genome Biol.
15
:
550
.
196.
Yu
G.
,
L. G.
Wang
,
Y.
Han
,
Q. Y.
He
.
2012
.
clusterProfiler: an R package for comparing biological themes among gene clusters.
OMICS
16
:
284
287
.

The authors have no financial conflicts of interest.