SUMMARY
Leukocyte diversity of the first-trimester maternal-fetal interface has been extensively described; however, the immunological landscape of the term decidua remains poorly understood. We therefore profiled human leukocytes from term decidua collected via scheduled cesarean delivery. Relative to the first trimester, our analyses show a shift from NK cells and macrophages to T cells and enhanced immune activation. Although circulating and decidual T cells are phenotypically distinct, they demonstrate significant clonotype sharing. We also report significant diversity within decidual macrophages, the frequency of which positively correlates with pregravid maternal body mass index. Interestingly, the ability of decidual macrophages to respond to bacterial ligands is reduced with pregravid obesity, suggestive of skewing toward immunoregulation as a possible mechanism to safeguard the fetus against excessive maternal inflammation. These findings are a resource for future studies investigating pathological conditions that compromise fetal health and reproductive success.
In brief
Sureshchandra et al. describe the changes in the decidual immune landscape in the third-trimester placenta compared with early pregnancy and the establishment of immunoregulatory mechanisms in decidual tissues as a mechanism of fetal protection from obesity-induced maternal inflammation.
Graphical abstract
INTRODUCTION
The placenta is a complex organ composed of maternal-derived decidua and fetal-derived chorionic villi.1 Functionally, it facilitates gas, nutrient, and waste exchanges between the fetus and the mother. The maternal compartment, called the decidua, harbors a unique immune landscape, which facilitates trophoblast invasion, maternal tolerance, debris clearance, and antimicrobial defense.2 Studying longitudinal changes in frequencies, phenotype, and function of decidual immune cells with healthy gestation is of great interest, given the role of decidua in the pathophysiology of several obstetric complications such as preterm labor,3 preeclampsia,4–6 and chorioamnionitis.7 First-trimester decidua (10–12 weeks) is composed mostly of decidual Natural Killer cells (dNKs, ~70% of immune cells), decidual macrophages (20%–25%), and T cells (3%–10%).8,9 The dNKs are the earliest immune cells detected in the decidua, differentiating from blood NK cells, and have been demonstrated to play a critical role in regulating trophoblast invasion and decidualization.10 Decidual macrophages and T cells play important roles in maintaining immune tolerance,11 mediating antimicrobial responses,2 promoting spiral artery remodeling,12 and clearing apoptotic bodies from the placental vasculature.13 While the diversity and characteristics of decidual immune cells during the first trimester have been well described, those at term remain poorly defined. Current models predict that over the course of healthy gestation, dNK and macrophage numbers dwindle while those of T cells remain intact in the decidua basalis.14 However, there is still no consensus about the phenotypic and functional diversity of decidual T cells and macrophages at term.
Maternal pre-pregnancy (pregravid) obesity is associated with an increased risk for several obstetric complications, including gestational diabetes and gestational hypertension,15,16 pre-eclampsia,17 placental abruption,18 preterm delivery,19,20 and need for cesarean delivery.21,22 These predispositions are mediated by altered placental structure,23,24 increased levels of oxidative stress,25 and elevated expression of proinflammatory factors.26 Studies in animal models suggest that changes in the decidual inflammatory environment can be attributed to dysregulated cell turnover and immune cell rewiring27,28; however, these changes have been poorly defined in humans. For example, first-trimester uterine NK cells in pregnant women with obesity exhibitan imbalance ofinhibitoryand activatingreceptors resulting in aberrant activation.29 Immunohistochemistry and flow cytometry-based studies have reported increased CD68+ macrophage frequencies in chorionic villi with maternal obesity.30 In line with these observations, higher transcript levels of proinflammatory genes IL6, CCL2, IL8, and TLR4 were detected in a baboon model of high-fat diet-induced obesity,31,32 suggesting potential immune cell adaptations with obesity at the maternal-fetal interface. However, a clear consensus on these observations is lacking, perhaps due to differences in choices of placental anatomic sections analyzed, immune markers used, experimental readouts measured, and confounding variables in human studies.23
A detailed understanding of immune cell adaptations in the decidua that support late pregnancy is currently lacking. More importantly, how maternal obesity reprograms the immune milieu of decidua at term is yet to be fully assessed. To address these gaps in our knowledge, we carried out a multi-pronged study using single-cell RNA sequencing (scRNA-seq), flow cytometry, Luminex, and functional assays to profile the diverse subsets of macrophages, T cells, and NK cells in term decidua and compared them with the single-cell atlas of early (6–12 weeks of gestation) maternal-fetal interface reported by Vento-Tomo et al.9 The use of decidua isolated from a cohort of pregnant mothers undergoing scheduled cesarean delivery in the absence of any obstetric complications allowed us to study the impact of pregravid obesity on immune cell adaptations at term without the confounders of labor.
Our data show that relative to the first trimester, decidual immune cells exhibit signs of heightened activation status at term. Furthermore, the relative abundance of T cells increases at term with a concomitant reduction in frequencies of macrophages and NK cells. We also report a shift from the dNK1 subset dominant at 12 weeks, to the dNK3 subset at term. At term, compared with their blood counterparts, decidual T cells exhibited enhanced regulatory and Th17 responses, whereas Th1 responses were attenuated, in line with immune tolerance. Finally, we observed two major macrophage subsets that can be distinguished based on HLA-DR and CD11c expression that may represent a tissue-resident and a monocyte-derived subset. Pregravid obesity led to a reduction of T cell numbers but increased accumulation of macrophages and functional rewiring of HLA-DRhighCD11chigh macrophages. Furthermore, decidual macrophages from mothers with obesity generated dampened responses to bacterial ligands, suggesting skewing toward regulatory phenotype. Collectively, our study provides an atlas of the immune landscape of term decidua and adaptations associated with pregravid obesity, shedding light on immune mechanisms limiting fetal exposure to maternal inflammation.
RESULTS
The immune cell landscape of decidua at term
While the diversity of human immune cells at the maternal-fetal interface during the first trimester has recently been described,9 the phenotypic and functional diversity of decidual immune cells at term is less understood. Here, we characterized CD45+ decidual leukocytes and matched peripheral blood mononuclear cells (PBMCs) obtained from pregnant women undergoing scheduled cesarean delivery using a combination of single-cell sequencing, flow cytometry, and functional assays (Figure 1A). Live CD45+ decidual leukocytes and PBMCs collected at term from four subjects were sorted and profiled using scRNA-seq (Figure 1A). Major decidual immune cell subsets were identified following dimensionality reduction and removal of potential blood contaminants following the integration of immune cells from blood and decidua (Figures S1A and S1B). The expression of structural genes defining tissue-resident macrophages (VIM) and T cells (ANXA1, EZR, TUBA1A) was higher in decidual leukocytes compared with PBMCs (Figure S1C), confirming their tissue-resident identity.
Figure 1. Defining the immune landscape of term decidua.
(A) Experimental design: CD45+ leukocytes were isolated from maternal blood and decidua from term cesarean deliveries (n = 4). Single-cell suspensions of decidual leukocytes and matched PBMCs were subjected to gene expression profiling using 10× 3′ single-cell gene expression protocol (scRNA-seq).
Additionally, sorted decidual and peripheral T cells were surface stained with DNA oligo-tagged antibodies and hashing antibodies and subjected to 5′ gene expression and TCR on the 10x platform (n = 5). Finally, the frequency, phenotype, and function of various immune cells were assayed using flow cytometry.
(B) Uniform Manifold Approximation and Projection (UMAP) of 12,698 CD45+ decidual leukocytes from four donors.
(C) Violin plots showing log-transformed normalized expression levels of major cell-type determining markers identified using Seurat.
(D) Stacked bar graph comparing frequencies of major immune cell types in term decidua (n = 62) using multiparameter flow cytometry. Error bars represent the mean and standard error of the mean (SEM).
(E) Changes in frequencies of major immune cell subsets in the decidua based on scRNA-seq obtained from first-trimester (T1, weeks 6–12) decidua9 integrated with week 37 (T3) data generated in this study.
(F) Feature plots comparing expression of transcription factors, surface markers, and cytokines associated with immune activation at T3 relative to T1.
Overall, the decidual immune landscape was dominated by T cells, NK cells, and macrophages (Figure 1B). Decidual CD4 and CD8 T cells were identified based on the relative expression of IL7R, CCR7, and XCL1, while regulatory T cells (Tregs) were identified based on the high expression of CTLA4 and FOXP3 (Figures 1B and 1C, Table S1). dNKs were identified based on the expression of NKG7, NCAM1, and CD160; macrophages based on the expression of CD14 and CD68; and dendritic cells (DC) based on the expression of FCER1A (Figure 1C and Table S1). Decidual immune cell distribution was validated using flow cytometry in a larger cohort of samples (n = 62) (Figures S1D and 1D).
We next integrated our data with those obtained from 6- to 12-week-old (first-trimester) decidua to understand longitudinal immune changes (Figures S1E and S1F). Compared with the first trimester, the relative frequencies of dNK cells and macrophages decreased while those of T cells, notably Tregs, increased at term (Figures 1E, S1E, and S1F). Moreover, both T and NK cells expressed high levels of NFKB and IFNΓ (Figure 1F), while decidual macrophages expressed higher levels of NFKB1 and NLRP3, CD83, and proinflammatory cytokines TNF and IL1B (Figure 1F), suggesting a state of heightened immune activation at term.
To eliminate the possibility of contaminating fetal cells in decidua, we integrated decidual leukocytes with matched chorionic villi and cord blood cells (n = 4) (Figures S2A–S2D). This analysis showed a lack of shared clusters with cord blood or contaminating Hofbauer cells (cluster 16) (Figure S2E), fetal cells found exclusively in the chorionic villous, in the decidua. Furthermore, monocytes were restricted to the cord blood mononuclear compartment (Figure S2E). This analysis confirms the clear separation of the decidua from the chorionic villous during manual dissection of the placenta at delivery and the lack of contaminating fetal cells in the decidua single-cell analysis.
Gestational shifts in dNK cells
We next investigated shifts in the dNK subsets with pregnancy. Within the dNK cell population, the frequency of dNK1 (expressing B4GALNT1 and CYP26A1), which is the largest cluster found at T1,9 was reduced at T3 (Figure S3A). Similarly, dNK2, a subset of cytotoxic dNKs expressing high levels of granulysin (GNLY) and granzymes (GZMB and GZMH) (Figures S3A and S3B) also decreased in proportion between T1 and T3. On the other hand, the frequency of dNK3 (expressing inhibitory receptors CD160 and KLRB1) increased at T3 relative to T1 (Figures S3A and S3B).
We then used flow cytometry to confirm and validate these shifts in dNK subsets. The dNK subsets were identified using CD103, CD49a, CD39, and ITGB2, as previously described,9 as well as activation markers NKG2A, NKG2C, KIR2DL4 (CD158d), KIR2DL2/3 (CD158b), KIR2DL1 (CD158a), and CD160 (Figure S3C). The frequencies of dNK2 and dNK3 were significantly higher than dNK1 at T3 (Figure S3D). Expression of activating (NKG2A) and inhibitory (NKG2C) receptors, as well as killer immunoglobulin receptor KIR2DL4, an activating receptor for HLA-G expressed on extravillous trophoblasts, were highest on dNK2 followed by dNK3 and lowest on dNK1 (Figures S3E and S3F). Expression of HLA-C receptors also varied among dNK subsets—a higher proportion of dNK3 cells expressed KIR2DL1, whereas KIR2DL2/3 expression was highest in dNK2 (Figure S3G). The inhibitory receptor CD160 was expressed at a lower level on dNK3 cells (Figure S3H). Finally, all dNK subsets were capable of degranulation as indicated by increased levels of CD107a in response to PMA/ionomycin stimulation (Figure S3I).
Phenotypic and clonal diversity of decidual T cells at term
In contrast to circulating T cells, decidual T cells were predominantly of effector memory phenotype (Figures 2A and S4A). To compare the phenotypic and clonal diversity of decidual T cells relative to blood T cells, we performed 5′ gene expression and T cell repertoire analyses at the single-cell level coupled with cell surface protein analysis using TotalSeq-C technology (Figure 1A, n = 4/group). Dimensionality reduction, clustering, and integration of blood and decidual T cells showed 32 clusters (Figure S4B). Naive T cell clusters were identified based on high expression of IL7R, SELL, and CCR7 (Figure S4C) and excluded from further analysis. The remaining 13 non-naive T cell clusters (red font) were reorganized into 17 clusters of memory T cells (Figures 2B and 2C, and Table S2). Interestingly, there was very little overlap between the decidual and blood compartments (Figure S4D), occurring only within proliferating, activated CD8, and CM CD4 T cell clusters (Figures 2B and S4D). Overall, decidual T cells expressed higher levels of CD69 and PD-1 (both at protein and gene expression levels) but lower levels of SELL (encoding CD62L) compared with blood memory T cells (Figures 2D, 2E, and S4E). Single-cell analyses also showed an increased abundance of regulatory T cells (expressing high levels of FOXP3 and CTLA4 and surface CD25) (Figures 2C and S4F), which was confirmed by flow cytometry (Figure 2F).
Figure 2. Phenotypic and clonal diversity of decidual T cells at term.
(A) Frequencies of naive and memory CD4 and CD8 T cell subsets in blood (n = 55) and decidua (n = 6) at T3 using flow cytometry. Error bars represent the mean and standard error of the mean (SEM).
(B) UMAP of matched decidual and blood memory T cells at term (n = 4), annotated by surface- and gene expression markers.
(C) Stacked violin plots of key gene markers of memory T cell subsets within clusters using normalized transcript counts. Red font indicates clusters shared between blood and decidual leukocytes.
(D) Frequencies of CD4 (top) and CD8 T cells (bottom) in blood (n = 3) and decidua (n = 9) expressing CD69 and CD103. Error bars represent the mean and standard error of the mean (SEM).
(E and F) Frequencies of (E) PD-1+ CD4 and CD8 T cells (n = 3/group) and (F) Tregs within CD4 (n = 3 for blood and n = 9 for decidua). Error bars represent the mean and standard error of the mean (SEM).
(G) Th1 cytokine responses of blood (n = 3) vs. decidual (n = 9) CD4 (left) and CD8 (right) T cells in response to 4 h PMA/ionomycin stimulation.
(H) Treg and Th17 cytokine responses of blood (n = 3) vs. decidual (n = 3) CD4 T cells following 4 h PMA/ionomycin stimulation.
(I) T cell clone sizes in blood and decidua at term.
(J) Chao diversity of T cell clones from blood and decidua within each donor.
(K) Clonal tracking of the 10 most abundant blood T cell clones in the decidua (top panel) and blood (bottom panel) in each donor. ****p < 0.0001, ***p < 0.001, **p < 0.01, and *p < 0.05.
Gene expression profiles suggest heightened activation of decidual T cells relative to those in circulation as indicated by increased expression of NFKB1, IFNΓ, and TNF (Figure S4G). However, Th1 cytokine production (tumor necrosis factor [TNF] α, macrophage inflammation protein [MIP]-1β, and interferon [IFN]γ) by decidual T cells following PMA/ionomycin stimulation was attenuated compared with circulating T cells (Figure 2G). Single-cell analyses also identified a Th17 (expressing RORA) cluster in the decidua (Figures 2B and 2C), which aligned with significantly higher IL-17 production by decidual compared with circulating T cells (Figure 2H). Finally, enhanced transforming growth factor (TGF)-β production following PMA/ionomycin stimulation correlated with higher frequencies of regulatory T cells (Figure 2H).
We next compared T cell receptor (TCR) repertoire diversity between blood and decidual memory T cells based on paired TCR alpha and beta genes captured in the single-cell assay (Figure 2B). Analysis of clonal sizes shows reduced TCR repertoire diversity in the decidua as indicated by the absence of large T cell clones (Figure 2I) and reduced Chao diversity (Figure 2J). Additionally, in all four donors, T cell clones that were predominant in the blood were detected in the decidua, albeit at lower proportions (Figure 2K, top panel). On the other hand, predominant decidual T cell clones were only detected in the blood of three of the four donors (Figure 2K, bottom panel), with proportions higher than in the blood in only one of the four donors.
Two distinct macrophage subsets exist in term decidua
Macrophages made up roughly 20% of leukocyte frequencies at term (compared with 28% in the first trimester) (Figure 1E). Within the decidua, we identified two major subsets of macrophages based on surface expression of HLA-DR and CD11c (Figures 3A and 3B), as well as size/granularity (Figure S5A). Both subsets had distinct transcriptional profiles compared with matched peripheral monocytes (Figure 3A). The subset expressing lower levels of HLA-DRA (HLA-DRlow) expressed higher levels of alarmins S100A8, S100A9, S100A10, IL1B, CXCL8, and markers associated with in vitro IFNγ conditioned macrophages (TREM1) (Figures 3A and 3B). On the other hand, the HLA-DRhigh subset expressed higher levels of tetraspanins (CD9 and CD81) and canonical markers associated with in vitro IL-4 conditioned macrophages (TREM2, MSR1, APOE) (Figures 3A and 3B). Both subsets of macrophages were also detected in first-trimester decidua (Figure S1E), with a modest increase in HLA-DRlow subset (22% at T1 vs. 34% at T3) and modest decrease in HLA- DRhigh subset (79% at T1 vs. 67% at T3) with gestational age. However, both subsets of macrophages were highly activated at term, with roughly 50%–75% of HLA-DRhigh macrophages expressing activation marker CD83, cytokine IL1B, and chemokine CXCL8 (Figure S5B). The presence of these two decidual macrophage subsets at term was confirmed by flow cytometry (Figure 3C) and imaging flow cytometry (Figure 3D) with the HLA-DRhigh subset expressing higher levels of CD11c compared with the HLA-DRlow subset.
Figure 3. Two distinct subsets of macrophages exist in term decidua.
(A) Heatmap of top marker genes for the decidual HLA-DRlow and HLA-DRhigh subsets, classical and non-classical blood monocytes. Each column is a bin of a fixed number of cells and each row represents a gene. A subset of genes within each cluster is annotated. High and low expression are represented as red and blue, respectively.
(B) Violin plots comparing expression of gene markers within decidual macrophage subsets.
(C) Surface expression of HLA-DR and CD11c measured by flow cytometry.
(D) Brightfield and fluorescence profiles of macrophage subsets captured using imaging flow cytometry.
(E) Median Fluorescence Intensities (MFI) of macrophage surface markers on decidual macrophage subsets (n = 7/group).
(F) Frequencies of TREM2+ FOLR2+ (n = 5/group) and CD163+ (n = 7/group) within decidual macrophages.
(G) Phospho p65 signal in blood monocytes and decidual macrophage subsets (n = 3/group) by flow cytometry. Error bars represent the mean and standard error of the mean (SEM).
(H) Comparison of baseline secreted cytokine profiles (median) of sorted decidual macrophage subsets (n = 4/group) and blood monocytes (n = 2). * and # represent significant differences relative to blood monocytes between HLA-DRlow and HLA-DRhigh macrophages, respectively.
(I) Responses to overnight lipopolysaccharide (LPS) stimulation of total decidual leukocytes (n = 8/group). Error bars represent the mean and standard error of the mean (SEM).
(J) MFI of intracellular cellROX (n = 7/group).
(K) Percentage of E. coli pHrodo+ (n = 8/group) in decidual macrophages.
(L) Oxygen consumption rate and extracellular acidification rates of decidual macrophages (n = 6/group). ****p < 0.0001, **p < 0.01, *p < 0.05, and #p<0.1.
To further characterize these decidual macrophage subsets at term, we measured protein expression of several markers associated with in vitro polarized macrophages (Figure S5C). Interestingly, surface expression of TREM-1 was lower on the HLA-DRlow subset despite the higher transcript levels. Moreover, surface expression of CCR2 was higher in the HLA-DRlow subset (Figure 3E), suggesting that this population may likely be infiltrating monocyte-derived macrophages. On the other hand, HLA-DRhigh macrophages expressed higher levels of surface markers CD86, CD11c, CD16, CD64, and TLR4 (Figures 3E, S5C, and S5D) that are traditionally associated with IFNγ conditioned macrophages. Interestingly, this subset was also enriched for regulatory CD163+ and TREM2+FOLR2+ macrophages (Figures 3E, 3F, and S5E), suggesting that this population may be a tissue-resident.
Functional diversity of macrophage subsets in term decidua
We next investigated if the transcriptional heterogeneity of decidual macrophages correlated with functional diversity. Flow analyses showed that the HLA-DRhigh macrophage subset exhibited a more heightened state of activation, expressing higher levels of phosphorylated p65 (Figure 3G). This heightened activation state was further evident when we assessed their baseline cytokine/chemokine secretion profiles. Sorted blood monocytes, and both macrophage subsets from the same donors were cultured in RPMI for 16 h, and their secretomes were analyzed using Luminex. Decidual macrophages secreted higher IL-12p70, IL-23, IL-10, CXCL9, S100B, granulocyte-macrophage colony-stimulating factor (GM-CSF), and CXCL11 compared with blood monocytes (Figure 3H). However, secreted levels of both TNFα and IL-10 were significantly higher in HLA-DRhigh relative to HLA-DRlow macrophages (Figure 3H), suggesting differential activation states between the two subsets.
Moreover, a higher percentage of HLA-DRhigh macrophages responded to lipopolysaccharide (LPS) stimulation as evidenced by the increased frequency of IL-6+TNFα+ producing cells (Figure 3I), expressed higher levels of reactive oxygen species (ROS) (Figures 3J and S6A), and were more phagocytic (Figure 3K). Since these functions are intricately linked to the metabolic state of the cell, we next compared several aspects of cellular metabolism. Our analysis shows that HLA-DRhigh macrophages are more lipid-laden than HLA-DRlow macrophages (Figure S6B). Moreover, seahorse energy phenotype assay indicated that at baseline, the HLA-DRhigh macrophages had a higher oxygen consumption rate (OCR) and lower extracellular acidification rate (ECAR) (Figure 3L) correlating with higher mitochondrial membrane potential (Figure S6C). Finally, HLA-DRhigh macrophages exhibited greater uptake of glucose analog 2-NBDG and increased expression of glucose transporter GLUT1 (Figure S6D). Collectively these observations suggest HLA-DRhigh macrophages are more metabolically active than HLA-DRlow cells.
Identification of heterogeneous macrophage subsets within HLA-DRhigh subset
Given the elevated expression of both inflammatory (TREM1) and regulatory markers (TREM2, CD163, FOLR2) within HLA-DRhigh macrophages (Figures 3E and 3F), we next investigated the heterogeneity of this subset. High-resolution clustering and visualization of Uniform Manifold Approximation and Projection (UMAP) showed three distinct macrophage clusters within HLA-DRhigh cells (Figure 4A) that were transcriptionally organized along a pseudo temporal trajectory originating from HLA-DRlow to cluster 1, cluster 2, and terminating in cluster 3 of HLA-DRhigh macrophages (Figures 4A and 4B). The genes defining the origin of this trajectory were primarily proinflammatory LYZ, NLRP3, TREM1, and S100 alarmins, which were highly expressed in cluster 1 (Figures 4C and 4D). Cells in cluster 2 expressed high levels of HLA-DRA, scavenger receptors (CD36, MARCO, MSR1), lipid signaling molecules (FABP5, CD9, CD63), and regulatory molecules (TREM2 and IL1RN) (Figures 4C and 4D). Finally, cells in cluster 3 express high levels of complement proteins (C1QA, C1QB), folate receptor FOLR2, and negative regulators of inflammation (ATF3, KLF2, KLF4) (Figures 4C and 4D). We then compared the transcriptomes of these clusters with recently published data of macrophages stimulated with TPP (TNF, Pam3CSK4, and prostaglandins PGE2), a model for chronic inflammation.33 In concordance with the trajectory analysis and the shift from proinflammatory to regulatory gene expression patterns, cluster 1 of HLA-DRhigh and HLA-DRlow macrophages had the highest scores while cluster 3 had the lowest score (Figure 4E).
Figure 4. Diverse subsets within HLA-DRhigh macrophages.
(A) UMAP highlighting diverse subsets of macrophages within HLA-DRhigh macrophages when analyzed under a higher resolution. Arrows point in the direction of pseudo temporal trajectory predicted by Monocle.
(B) Trajectory of cells ordered by predicted pseudo time colored by pseudotime (top) or by the cluster of origin (bottom).
(C) Heatmap of top 100 genes that define the trajectory originating from HLA-DRlow macrophages, through clusters 1, 2, and 3 of HLA-DRhigh macrophages. Low and high gene expression is indicated by blue and red, respectively.
(D) Violin plots comparing the expression of the top eight markers from each of the HLA-DRhigh macrophages across all three clusters.
(E) Violin plot comparing aggregate score for modules of genes highly expressed in macrophages stimulated with TPP (TNF+Pam3CSK4+PGE2), an in vitro model for chronic inflammation.
(F) Gating strategy for flow cytometry-based identification of HLA-DRhigh macrophage clusters based on markers discovered from differential gene analyses.
(G) Three-way functional enrichment of top genes expressed by each HLA-DRhigh subset performed in Metascape. The size of the bubble denotes the number of genes mapping to each gene ontology (GO) term, and the intensity of color denotes the statistical strength of prediction.
Canonical markers that can differentiate the three HLA-DRhigh subclusters were validated using intracellular staining of S100A8/A9, and surface expression of FOLR2 and CD9 by flow cytometry (Figure 4F). Functional enrichment of the top 100 marker genes from each sub-cluster showed that cluster 1 has a dominant signature associated with LPS response, while cluster 2 had features associated with wound healing and antigen presentation, and finally cluster 3 was enriched in regulatory pathways (Figure 4G).
Pregravid obesity is associated with reduced frequency of decidual T cells and functional rewiring of HLA-DRhigh decidual macrophages
The prevalence of pregravid obesity is increasing34 and is implicated in elevated risk for perinatal complications, which may have a long-term impact on maternal and offspring health.15,22,35–41 Therefore, we next investigated the impact of maternal pregravid obesity on the immunological landscape of the decidua. Frequency of major decidual immune subsets from lean pregnant women (BMI <25) was compared with those with obesity (BMI >30). Obesity had no impact on dNK cell frequencies (Figure 5A). In contrast, the proportions of CD4 and CD8 T cells within the CD45 compartment were significantly reduced (Figure 5A). Moreover, decidual CD4 and CD8 T cell frequencies negatively correlated with maternal pregravid BMI (Figure S7A). However, no differences in the relative abundance of naive/memory T cell subsets (Figure S7B) or tissue-resident subsets (Figure S7C) were observed with pregravid obesity. Furthermore, T cell responses to PMA/ionomycin stimulation were comparable between the two groups (Figure S7D). Regulatory T cell frequencies, on the other hand, increased with pregravid obesity (Figure 5A).
Figure 5. Decidual immune cell composition with maternal obesity.
(A and B) Dot plots comparing (A) lymphocyte and (B) macrophage proportions within CD45+ decidual cells in lean pregnant women (n = 6 for Tregs, 32 for rest) and those with obesity (n = 6 for Tregs, 31 for rest).
(C) Gating strategy and dot plots comparing frequencies of S100A9+ cells within HLA-DRhigh macrophages (n = 5/group). Error bars represent the mean and standard error of the mean (SEM).
(D) Stacked bar graph comparing frequencies of decidual leukocytes within total cells sequenced in each group (within CD45+ cells).
(E) Stacked bar comparing HLA-DRhigh macrophage cluster within total CD45+ cells.
(F and G) Gene Ontology (GO) terms of genes that are up- (F) and downregulated (G) with pregravid obesity within HLA-DRhigh macrophages predicted by Metascape. Only differentially expressed genes with a Log2 fold change cutoff of 0.4 were analyzed.
(H) Violin plots compared normalized transcripts of select genes that are significantly up- (left) or downregulated (right) with obesity.
(I) Dot plots comparing frequencies of TREM1+ cells or cytokine expressing cells within HLA-DRhigh macrophages (n = 5/group). Error bars represent the mean and standard error of the mean (SEM).
(J) Violin plots comparing secreted factors at baseline by FACS sorted HLA-DRhigh macrophages (n = 3/group).
(K) Experimental design for measuring ex vivo macrophage responses to E. coli or bacterial TLRs.
(L) Bar graphs comparing cytokine-producing cells TNFα+IL-6+ (left) and TNFα+IL-6+IL1β+ (right) following stimulation with bacterial TLRs and E. coli (n = 6/group). Error bars represent the mean and standard error of the mean (SEM). ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05.
Reduction in decidual T cells with pregravid obesity was accompanied by an expansion of decidual macrophages (Figure S7E), driven by the HLA-DRhigh subset (Figure 5B). Moreover, the frequency of HLA-DRhigh but not HLA-DRlow subsets positively correlated with maternal pregravid BMI (Figure S7F). Flow analyses of HLA-DRhigh macrophages showed an increase in S100A9+ macrophages (Figure 5C), a marker of subclusters 1 and 2 (Figure 4F), and recently infiltrated macrophages.42
Next, we leveraged scRNA-seq to further interrogate the impact of pregravid obesity on the transcriptional landscape of decidual leukocytes (Figure S8A). In line with the flow cytometry data, this analysis showed a contraction of CD4 and CD8 T cells and expansion of decidual macrophages (Figure 5D), driven by increased frequencies of all three HLA-DRhigh subsets (Figure 5E). Differential gene expression analyses of HLA-DRhigh macrophages between lean and obese groups showed an up-regulation of pathways associated with response to lipoproteins and cholesterol transport (FABP5, CD36, APOE, APOC1, CD9), response to stress (CEBPA, CEBPB, HSPA5, HSPA8), and antigen processing and presentation (HLA-DRA) (Figures 5F and 5H left column, Figure S8B left column, and Table S3). Proinflammatory signatures, on the other hand, were downregulated with pregravid obesity (Figure 5G and Table S3). This included cytokine signaling pathways (CCL4, CXCL8, TNF, NFKB1, REL), interferon-stimulated genes (ISG15, IFITM3), metabolic genes (COX6A1, COX6B1), and genes important for T cell activation (CD55, CD300E) (Figures 5G and 5H right column, and Figure S8B right column).
To validate the transcriptional changes described above, we assessed the protein expression of inflammatory mediators and markers using flow cytometry. While frequencies of TREM1+, MIP-1β+, and CXCL8+ decidual macrophages were reduced with pregravid obesity (Figure 5I), the frequency of IL-1RA+ macrophages increased (Figure 5I). Additionally, spontaneous production of several proinflammatory cytokines (TNFα, IL-1β, IL-12p70) was attenuated within sorted HLA-DRhigh macrophages (Figures 5J and S8C) in the absence of changes in baseline levels of activation-associated signaling molecules (Figure S8D). To define the biological significance of the downregulation of genes important for antimicrobial function with pregravid obesity (Figure 5G), we measured the frequency of cytokine-producing decidual macrophages following stimulation with a cocktail of bacterial TLR ligands (activating TLRs 1/2/4/6) or E. coli using flow cytometry (Figures 5K and S8E). Pregravid obesity was associated with reduced frequency of TNFα+IL-6+IL-1β+ macrophages following both stimulation conditions within both HLA-DRhigh (Figure 5L) and HLA-DRlow subsets (Figure S8F).
DISCUSSION
Roughly 30%–40% of decidual cells are immune cells that play a critical role in the maintenance of fetomaternal homeostasis.2,43 The phenotype and frequency of immune cells in the decidua dynamically adapt to fetal needs,44,45 either by proliferating locally or via recruitment from circulation.46 These time-sensitive changes support fetal growth and tolerance, protection from infections, and ultimately promote parturition.43,47 Recent advances in single-cell technologies have facilitated the unbiased profiling of rare populations of immune cells residing in the early decidua.9,48 While the transcriptional profiles of placental trophoblast populations from chorionic villi in homeostasis49,50 and in preterm labor have been recently investigated,51 an atlas of immune cell diversity of human decidua at term is still lacking. The availability of such a high-resolution map of decidual immune cells collected in the absence of pregnancy complications would pave the way for future research on the role of decidual T cells, NK cells, and macrophages in the pathophysiology of adverse outcomes such as preeclampsia, spontaneous abortion, and preterm labor. We, therefore, profiled the immune compartment of term decidua isolated from mothers undergoing scheduled cesarean delivery to generate a detailed map of transcriptomic, phenotypic, and functional diversity of term decidual T cells, NK cells, and macrophages. Finally, given the strong association between maternal pre-pregnancy BMI and pregnancy complications,3–7 we investigated the impact of pregravid obesity on the functional rewiring of decidual immune cells.
The first-trimester decidua is populated by NK cells (dNKs),52,53 which account for about 70% of decidual leukocytes that are recruited from blood by factors such as IL-15 and progesterone.54,55 Early dNKs facilitate embryonic development and promote decidual tolerance to the embryo. Furthermore, animal studies have shown the critical role of dNK-derived factors (vascular endothelial growth factor C [VEGF-C], TGF-β, IFNγ) in uterine vascular modifications and extravillous trophoblast (EVT) differentiation.56,57 scRNA-seq and mass cytometry analyses of first-trimester decidua9,10 showed three subpopulations of dNKs. Of the three populations, dNK1s (Eomes+) are the most abundant, expressing granzymes, KIR inhibitory receptors, and LILRB1 suggesting their preferential interactions with EVTs. This subset of dNKs produce growth factors such as pleiotrophin and osteoglycin, promoting fetal growth in both mice and humans,58 suggesting their critical role in early pregnancy. On the other hand, mass cytometry analyses suggest that dNK3s are predominantly Tbet+ and mounted the strongest polyfunctional (CD107a, MIP-1α, MIP-1β, XCL1, IFNγ) response to ex vivo PMA stimulation.10 Interestingly, these populations have also been described in the endometrium (uterine NKs [uNKs]) of non-pregnant women.59 Our analyses show a sharp drop in total dNK proportions at term, with a substantial shift toward dNK2 and dNK3 cells, as recently described in NK cells within the uterine mucosa (uNKs).59 This reduction is in line with the reduced need for vascular remodeling and a shift toward fetal growth and enhanced immune activation associated with labor in the third trimester.60 It is known that dNKs at term are transcriptionally distinct compared with their first-trimester counterparts and exhibit a lower capacity to respond to cytomegalovirus (CMV).61 Furthermore, dNKs become less granular over the course of a healthy pregnancy.62 However, their transcriptional diversity and activation status in response to labor, parturition, and adverse pregnancy outcomes associated with late-stage pregnancies is still unclear.
Reduction and redistribution of dNK cell subsets at term was associated with an expansion of T cells, potentially indicating a shift from tissue remodeling to host defense at the maternal-fetal interface. These observations are in line with recent studies that have used imaging mass cytometry to analyze immune cells in decidua in situ.63 T cells are recruited into the decidua from the blood by trophoblast-derived CXCL16.64 Tissue-resident memory (TRM, CD69+) CD8 T cells are present in term decidua, and a subset of these cells expressed CD103.65–67 TRM T cells have been reported to express high levels of PD-1 but low levels of effector molecules such as GZMB and perforin compared with their blood counterparts, in line with their ability to trigger trophoblast antigen-specific tolerance.68,69 We also observed increased PD-1 expression on decidual CD4 T cells; however, these TRMs were predominantly CD103–. Aberrant Th1 responses in placental CD4 T cells have been associated with pathologies such as recurrent spontaneous abortions.70 Our data indicate reduced ability of decidual CD8 T cells to produce IFNγ, MIP-1β, and TNFα compared with circulating CD8 T cells. While decidual and circulating CD4 T cell IFNγ responses were comparable, production of MIP-1β and TNFα were lower in decidual CD4 T cells compared with their blood counterparts. Th17 responses were significantly higher in the decidua. Both Th1 and Th17 responses at the maternal-fetal interface have been shown to increase over the course of pregnancy and play a role in the initiation of labor.71 Finally, CD4 T cells generated higher TGF-β responses in line with the increased frequency of Tregs, which play an important role in maintaining fetal tolerance and preventing miscarriages.72
This study compared the clonality of matched decidual and blood T cells. In all four donors, we observed that the decidual T cells repertoire was more restricted. However, in all four donors, the dominant decidual T cell clones were also detected in the blood, suggesting their recruitment from circulation. The process by which these specific clones are recruited and retained in the decidua, as well as their antigen specificity, remains poorly understood. Studies have shown that decidual CD4 and CD8 T cells can recognize viral antigens during Zika and dengue infections73 and that frequency of Epstein-Barr virus and CMV-specific CD8 T cells is greater in the decidua compared with blood.74 These findings suggest that decidual T cells can mediate antimicrobial responses.
Compared with decidual lymphocytes, macrophage frequencies modestly decreased at term relative to first-trimester decidua. A large macrophage population with the near absence of DCs suggests their potential role in antigen presentation at the maternal-fetal interface. Additionally, these cells play a critical role in regulating T cell responses, tissue remodeling, and clearance of apoptotic cells. Two distinct subsets of macrophages have been defined in decidua during early gestation—characterized by differential surface expression of CD11c and CCR2.75 Gene expression profiling of these subsets suggested that the CD11clow subset plays a role in tissue homeostasis while CD11chigh is important for antigen presentation.76 However, neither subset conformed with the in vitro derived IFNγ or IL-4 conditioned macrophages, suggesting an underappreciation of their plasticity at the maternal-fetal interface. Using unbiased scRNA-seq of matched term decidual macrophages and blood monocytes, we report two major macrophage subsets characterized by co-expression of HLA-DR and CD11c as described for first-trimester decidua.76
Trajectory analysis, flow cytometry, and profiling of secreted cytokines at baseline showed that the HLA-DRlow macrophages are an inflammatory subset that are transcriptionally similar to blood monocytes. On the other hand, despite high expression level of regulatory molecules (TREM2, CD163, CD206), HLA-DRhigh macrophages exhibited an enhanced capacity for TLR4-mediated responses and phagocytosis. Furthermore, HLA-DRhigh macrophages also accumulated higher levels of lipids and are metabolically primed for oxidative phosphorylation, supporting their identity as bona fide tissue-resident macrophages, as previously described in other compartments.77 Within the decidua, these macrophages have been shown to be more likely to be located within the vicinity of T cells at term.63 Further characterization of the heterogeneity within this subset showed three subpopulations predicted to play a role in acute inflammatory responses and killing (S100A8+ CD9−), response to wounding (S100A8+ CD9+), and apoptosis and fat-cell differentiation (S100A8− FOLR2+). Therefore, decidual macrophages span the spectrum from inflammatory blood-derived HLA-DRlow cells to HLA-DRhigh FOLR2+ regulatory cells involved in tissue remodeling and clearance of cellular debris.
A consistent theme across both decidual lymphocyte and macrophage populations was a heightened state of immune activation at term compared with the first trimester. This is evident from increased levels of inflammatory cytokine transcripts (IL1B, IFNΓ, TNF) across dNK, T cell, and macrophage subsets and higher levels of spontaneous secretion of cytokines and chemokines by sorted macrophage subsets compared with blood monocytes. We posit that this state of activation is important for parturition78 and is in line with recent studies highlighting progressive peripheral immune activation associated with gestation.79,80 Understanding these pregnancy-associated shifts in immune states is therefore critical in addressing the immunological basis of pregnancy complications.
Our analyses suggest a dramatic drop in CD4 and CD8 T cell numbers and concomitant increases in macrophage frequencies with pregravid obesity driven in a BMI-dependent manner by increased frequency of the HLA-DRhigh macrophage subset. Despite the overall loss of T cells, we report no preferential loss of any major T cell subset or functional loss in cytokine responses. Regulatory T cell frequencies, however, increased with obesity. In contrast, macrophage responses were significantly attenuated—with the downregulation of genes involved in pathogen sensing and cytokine signaling. Functionally, this translated to reduced levels of secreted inflammatory cytokines at baseline (TNFα, IL-1β) but also poor responses to ex vivo E. coli stimulation. We have previously demonstrated that pregravid obesity alters activation in peripheral monocytes at term, resulting in dampened ex vivo cytokine responses to TLR4 stimulation.22 These observations suggest continuous recruitment of circulation monocytes to the decidua.22,81–83
These observations align with previous reports of reduced CD163-macrophages in decidua parietalis of mothers with obesity.84 Taken together, these findings highlight an establishment of a tolerance-like program in term decidual macrophages with maternal obesity, mirroring the phenotype observed in peripheral monocytes reported in our previous study.22 Additionally, we posit that the accumulation of regulatory macrophages and Tregs with obesity is mediated by changes in the decidual microenvironment (cytokine/chemokine changes, stromal cell crosstalk),85 to protect the developing fetus from the undesirable effects of maternal inflammation. Suboptimal activation and attenuated responses can have untoward consequences for both host defense and initiation of labor. These changes could also contribute to obesity-associated pathologies during the late stages of pregnancy.
Limitations of the study
Our study is limited by restricting analyses to decidua collected following cesarean deliveries and the lack of investigating dietary contributions to maternal obesity-associated decidual adaptations. Moreover, while pregravid obesity has been shown to attenuate cord blood monocyte and T cell responses, its influence on fetal macrophage populations within the chorionic villi is still unknown. Future studies need to focus on identifying key immunological changes occurring in the placenta stratified by mode of delivery. Animal models of maternal obesity could be leveraged to fully elucidate longitudinal changes in vascularization, tissue remodeling, and immune activation in the decidua.
STAR★METHODS
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Dr. Ilhem Messaoudi (ilhem.messaoudi@uky.edu).
Materials availability
This study did not generate new unique reagents.
Data and code availability
The datasets supporting the conclusions of this article are available on NCBI’s Sequence Read Archive and project ID’s are listed in the key resources table.
This paper does not report original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
KEY RESOURCES TABLE
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
| ||
Antibodies | ||
| ||
CD3-FITC | BD Biosciences | Cat#556611; RRID:AB_396484 Clone-SP34 |
CD20-BV510 | BioLegend | Cat#302340; RRID:AB_2561941 Clone-2H7 |
CD56-BV711 | BioLegend | Cat#318336; RRID:AB_2562417 Clone-HCD56 |
CD16-PB | BioLegend | Cat#302021; RRID:AB_492978 Clone-3G8 |
CD14-AF700 | BioLegend | Cat#301822; RRID:AB_493747 Clone-M5E2 |
HLA-DR-APC-Cy7 | BioLegend | Cat#307618; RRID:AB_493586 Clone-L243 |
CD11c-APC | BioLegend | Cat#301614; RRID:AB_493023 Clone-3.9 |
CD123-PCP-Cy5.5 | BioLegend | Cat#306016; RRID:AB_2264693 Clone-6H6 |
CD86-BV605 | BioLegend | Cat#305430; RRID:AB_2563824 Clone-IT2.2 |
CD4-FITC | BioLegend | Cat#300538; RRID:AB_2562052 Clone-RPA-T4 |
CD4-PE | BioLegend | Cat#317410; RRID:AB_571955 Clone-OKT4 |
CD8b-ECD | Beckman Coulter | Cat#6607123; RRID:AB_1575983 Clone-2ST8.5H7 |
CD45RA-PerCP-Cy5.5 | Thermo Fisher Scientific | Cat#45-0458-42; RRID:AB_10718536 Clone-HI100 |
CCR7-PE-Cy7 | BioLegend | Cat#353226; RRID:AB_11126145 Clone-GO43H7 |
CD69-PE | BioLegend | Cat#310906; RRID:AB_314841 Clone-FN50 |
CD103-BV605 | BioLegend | Cat#350218; RRID:AB_2564283 Clone-BER Act1 |
CTLA4-PeDazzle594 | BioLegend | Cat#369616; RRID:AB_2632878 Clone-BNI3 |
CD25-APC-Cy7 | BioLegend | Cat#302614; RRID:AB_314284 Clone-BC96 |
FOXP3-A488 | BioLegend | Cat#320106; RRID:AB_439752 Clone-206D |
CD49a-PE-Cy7 | BioLegend | Cat#328312; RRID:AB_2566272 Clone-TS2/7 |
CD39-BV510 | BioLegend | Cat#328219; RRID:AB_2563265 Clone-A1 |
ITGB2 (CD11a/CD18)-PCP-Cy5.5 | BioLegend | Cat#302120; RRID:AB_2565587 Clone-TS1/18 |
KIR2DL4 (CD158d)-APC | BioLegend | Cat#347008; RRID:AB_2130691 Clone-mAb33 |
KIR2DL1 (CD158a)-APC-Cy7 | Novus Biologicals | Cat#NB100-63267APCCY7; RRID:AB_2941073 Clone-NKVFS1 |
KIR2DL2/3 (CD158b)-PE | BD Biosciences | Cat#559785; RRID:AB_397326 Clone-CH-L |
CD160-AF700 | Novus Biologicals | Cat#NBP2-90074AF700; RRID:AB_2941072 Clone-275 |
NKG2A-PE-Dazzle594 | BioLegend | Cat#375121; RRID:AB_2888869 Clone-S19004C |
NKG2C-PE | BioLegend | Cat#375003; RRID:AB_2888871 Clone-S19005E |
CD9-PE-Cy7 | BioLegend | Cat#312116; RRID:AB_2728256 Clone-HI9a |
CD68-APC | BioLegend | Cat#333810; RRID:AB_2275735 Clone-Y1/82A |
TLR4-BV711 | BD Biosciences | Cat#564404; RRID:AB_2738794 Clone-tf901 |
CD163-APC/Fire750 | BioLegend | Cat#333634; RRID:AB_2734333 Clone-GHI/61 |
CD206-BV510 | BioLegend | Cat#321138; RRID:AB_2721530 Clone-15-2 |
CD209-FITC | BioLegend | Cat#330104; RRID:AB_1134048 Clone-9e9a8 |
TREM1-AF405 | R&D Systems | Cat#FAB12781V; RRID:AB_2941074 Clone-888111 |
TREM2-PerCP | R&D Systems | Cat#FAB17291C; RRID:AB_2941075 Clone-237920 |
CD64-BV711 | BioLegend | Cat#305042; RRID:AB_2800778 Clone-10.1 |
CD64-APC | BioLegend | Cat#305014; RRID:AB_1595428 Clone-10.1 |
GLUT1-AF488 | R&D Systems | Cat#FAB1418G; RRID:AB_2941076 Clone-202915 |
FOLR2-PE | BioLegend | Cat#391704; RRID:AB_2721336 Clone-94b/folr2 |
CCR2-BV605 | BioLegend | Cat#357213; RRID:AB_2562702 Clone-K036C2 |
CD19-PE | BioLegend | Cat#302208; RRID:AB_314238 Clone-HIB19 |
S100A9-APC | Invitrogen | Cat#MA5-28129; RRID:AB_2745112 Clone-10.1 |
CD107a-FITC | BD Biosciences | Cat#555800; RRID:AB_396134 Clone-H4A3 |
PD-1-BV510 | BioLegend | Cat#329932; RRID:AB_2562256 Clone-Eh12.2h7 |
CD45-FITC | BioLegend | Cat#304038; RRID:AB_2562050 Clone-HI30 |
TNFα-APC | BioLegend | Cat#502912; RRID:AB_315264 Clone-Mab11 |
IL-6-FITC | BD Biosciences | Cat#554696; RRID:AB_395514 Clone-MQ2-6A3 |
IL-1β-PB | BioLegend | Cat#511710; RRID:AB_2124350 Clone-H1b-98 |
IFNγ-APC | Invitrogen | Cat#17-7319-82; RRID:AB_469506 Clone-4S.B3 |
IFNγ-PE-Cy7 | eBioscience | Cat#502527; RRID:AB_1626154 Clone-4S.B3 |
IL-2-AF700 | BioLegend | Cat#500320; RRID:AB_528929 Clone-MQ-17H12 |
IL-4-APC | Tonbo Biosciences | Cat#50-210-2786; RRID:AB_2941077 Clone-MP425D2 |
IL-17-FITC | Invitrogen | Cat#11-717942; RRID:AB_10805390 Clone-Ebio64dec17 |
TGF-β-PerCP-Cy5.5 | BioLegend | Cat#141410; RRID:AB_2561592 Clone-TW7-16B4 |
MIP-1β-PE | BD Biosciences | Cat#550078; RRID:AB_393549 Clone-D21-1351 |
GZMB-AF700 | BD Biosciences | Cat#560213; RRID:AB_1645453 Clone-GB11 |
NF-κB p50-AF488 | Luminex | Cat#4700-1674; RRID:AB_2941078 |
7-Amino-Actinomycin D (7-AAD) | Luminex | Cat#400-0290; RRID:AB_2941079 |
NF-κB p65 (pS529)-AF647 | BD Biosciences | Cat#558422; RRID:AB_647136 Clone-K10-895.12.50 |
IκBa-PE | eBioscience | Cat#12-903642; RRID:AB_2572683 Clone-MFRDTRK |
Phospho-p38 MAPK-APC | eBiosceince | Cat#17-9078-42; RRID:AB_2573290 Clone-4NIT4KK |
CD3-PE | BD Biosciences | Cat#556612; RRID:AB_396485 Clone-SP34 |
TotalSeq-C0072-CD4 | BioLegend | Cat#300567; RRID:AB_2800725 Clone-RPA-T4 |
TotalSeq-C0046-CD8 | BioLegend | Cat#344753; RRID:AB_2800922 Clone-SK1 |
TotalSeq-C0148-CCR7 | BioLegend | Cat#353251; RRID:AB_2800943 Clone-G043H7 |
TotalSeq-C0063-CD45RA | BioLegend | Cat#304163; RRID:AB_2800764 Clone-HI100 |
TotalSeq-C0146-CD69 | BioLegend | Cat#310951; RRID:AB_2800810 Clone-FN50 |
TotalSeq-C0145-CD103 | BioLegend | Cat#350233; RRID:AB_2800933 Clone-Ber-ACT8 |
TotalSeq-C0088-PD-1 | BioLegend | Cat#329963; RRID:AB_2800862 Clone-EH12.2H7 |
TotalSeq-C0085-CD25 | BioLegend | Cat#302649; RRID:AB_2800745 Clone-BC96 |
TotalSeq-C0251 | BioLegend | Cat#394661; RRID:AB_2801031 Clone-LNH-94, 2M2 |
TotalSeq-C0254 | BioLegend | Cat#394667; RRID:AB_2801034 Clone-LNH-94, 2M2 |
TotalSeq-C0256 | BioLegend | Cat#394671; RRID:AB_2820042 Clone-LNH-94, 2M2 |
TotalSeq-C0260 | BioLegend | Cat#394679; RRID:AB_2820046 Clone-LNH-94, 2M2 |
| ||
Biological Samples | ||
| ||
Fetal Bovine Serum, USDA Certified, Heat Inactivated | Omega Scientific | Cat#FB-02 |
FetalPlex™ Animal Serum Complex | GeminiBioProducts | Cat#100-602 |
| ||
Chemicals, Peptides, and Recombinant Proteins | ||
| ||
RPMI 1640 | VWR | Cat#45000-396 |
Hanks Balanced Salt Solution (HBSS) | VWR | Cat#21-023-CV |
Penicillin Streptomycin | GeminiBio | Cat#400109 |
L-Glutamine | GeminiBio | Cat#400106 |
N-2-hydroxyethylpiperazine-N′2-ethanesulfonic acid HEPES | ThermoFisher | Cat#15630080 |
Collagenase | Sigma | Cat# C9722 Source: Clostridium histolyticum |
Ammonium chloride - NH4Cl (RBC Lysis Buffer) | Sigma | Cat#A9434 |
Sodium hydrogen carbonate - CHNaO3 (RBC Lysis Buffer) | Fisher | Cat#AAA170050E |
EDTA (RBC Lysis Buffer) | Invitrogen | Cat#AM9260G |
Percoll Density Gradient | Neta Scientific | Cat# 17-0891-01 |
Ficoll-Paque | GE Healthcare | Cat# 17144003 |
Dimethyl sulfoxide (DMSO) | Sigma | Cat# D2650-100ML |
FOXP3 Fix/Perm Buffer Set | BioLegend | Cat#421403 |
Ghost Dye 510 | TONBO Biosciences | Cat#13-0870-T 100 |
Ghost Dye 540 | TONBO Biosciences | Cat#13-0879 |
SYTOX Blue Dead Cell Stain | ThermoFisher Scientific | Cat#S34857 |
Human TruStain FcX | BioLegend | Cat#422302 |
Human True-Stain Monocyte Blocker | BioLegend | Cat#426103 |
Lipopolysaccharide (LPS) TLR4 ligand E. coli 055:B5 | Invivogen | Cat#tlrl-b5lps |
Pam3CSK4 TLR1/2 agonist | Invivogen | Cat#tlrl-pm2s-1 |
FSL-1 TLR2/6 agonist | Sigma | Cat#SML1420 |
E. coli | Migula. Castellani and Chalmers ATCC 11775 | Cat#1132342 |
Phorbol myristate acetate-NF-κB Activator (PMA) | Invivogen | Cat#tlrl-pma |
Ionomycin | Invivogen | Cat#inh-ion |
Brefeldin A (BFA) | BioLegend | Ca#420601 |
Fixation buffer | BioLegend | Cat#420801 |
Amnis NF-κB translocation kit | Luminex Corporation | Cat #ACS10000 |
Permeabilization wash buffer | BioLegend | Cat#421002 |
Bovine Serum Albumin (BSA), Fraction V— Molecular Biology Grade |
GeminiBio | Cat#700-106P |
Cytofix fixation buffer | BD Biosciences | Cat#554655 |
BD Perm/Wash buffer | BD Biosciences | Cat#554723 |
| ||
Critical Commercial Assays | ||
| ||
BODIPY 493/503 | ThermoFisher Scientific | Cat#D3922 |
pHrodo Deep Red E. coli BioParticle Conjugates | ThermoFisher Scientific | Cat#P35360 |
CellROX Deep Red Flow Cytometry Assay Kit | ThermoFisher Scientific | Cat#C10491 |
2-NBDG | ThermoFisher Scientific | Cat#N13195 |
MitoTracker Red CMXRos | ThermoFisher Scientific | Cat#M7512 |
Seahorse XF Cell Energy Phenotype kit | Agilent Technologies | Cat#103592-100 |
Human Premixed 29-plex Magnetic Luminex Assay | R&D Systems | Cat#Custom-LxSA-H-29 Lot#C0003514 |
TotalSeq B Hashtag Oligos 1-10 | BioLegend | 384631-392649 |
Chromium Single Cell 3’ Reagent Kits v3 | 10X Genomics | PN-1000075 |
3′ Feature Barcode Kit | 10X Genomics | PN-1000262 |
Chromium Single Cell 5’ Reagent Kits V2 | 10X Genomics | PN-1000263 |
5′ Feature Barcode Kit | 10X Genomics | PN-1000256 |
| ||
Deposited Data | ||
| ||
3′ Single Cell GEX data - Decidua | This paper | NCBI Sequence Read Archive: PRJNA817521 |
5′ Single Cell TCR data - Decidua | This paper | NCBI Sequence Read Archive: PRJNA817521 |
5′ Single Cell TCR data - Maternal PBMC | This paper | NCBI Sequence Read Archive: PRJNA817521 |
3′ Single Cell GEX data -Villous | This paper | NCBI Sequence Read Archive:PRJNA946160 |
3′ Single Cell GEX data - UCBMC | Sureshchandra et al. 202166 10.3389/fimmu.2021.617592 |
NCBI Sequence Read Archive:PRJNA690128 |
3′ Single Cell GEX data - Maternal PBMC | This paper and Sureshchandra etal. 202166 10.1016/j.isci.2021.102690 |
NCBI Sequence Read Archive:PRJNA887020 |
| ||
Software and Algorithms | ||
| ||
Prism | GraphPad | Version#8 |
Immunarch | R package | Version#0.6.7 |
Metascape | www.metascape.org | N/A |
Attune NxT Flow Cytometer Software | ThermoFisher Scientific | Version#2.5 |
FlowJo | TreeStar | Version#10.5 |
Ideas Analysis Software | Luminex Corporation | Version#6.1 |
Seahorse Wave | Agilent Technologies | Version#2.6.1 |
xPONENT® Software for Luminex Instruments | Luminex Corporation | Version#4.3 |
Cell Ranger Single-Cell Software Suite | 10X Genomics | Version 6.0.2 |
Seurat | R package | Version 3.1.5 |
Monocle | R package | Version 2.8.0 |
EXPERIMENTAL MODEL AND SUBJECT PARTICIPANT DETAILS
This study was approved by the Institutional Ethics Review Board of Oregon Health & Science University and the University of California, Irvine. A total of 102 non-smoking women (47 lean, 12 overweight, and 43 obese) who had an uncomplicated pregnancy were enrolled for this study. Cohort characteristics are outlined in Table 1. Written informed consent was obtained from all participants. Due to the strong positive correlation between pre-pregnancy BMI and total body fat in our previous studies,86 we used pre-pregnancy BMI as a surrogate for maternal pregravid obesity and stratified participants into lean or obese based on pre-pregnancy BMI (Table 1). Participants Only decidua from patients who met study inclusion criteria undergoing scheduled cesarean deliveries for a variety of medical and elective indications, including repeat cesareans, breech presentation, maternal request, and medical contra-indication to vaginal birth. There were no indications of complications for any of the study participants. The decision to use decidua obtained via cesarean section was made to avoid confounders associated with delivery (length of labor, methods of pain control, etc.). Exclusion criteria include active maternal infection, documented fetal congenital anomalies, substance use disorder, chronic illness requiring regular medication use, preeclampsia, gestational diabetes, chorioamnionitis, and significant medical conditions (active cancers, cardiac, renal, hepatic, or pulmonary diseases).
Table 1.
Cohort characteristics
Lean | Overweight | Obese | |
---|---|---|---|
| |||
Enrollment | 47 | 12 | 43 |
BMI (kg/m2)a | 21.9 (1.7) | 27.4 (1.6) | 38 (7.9) |
Maternal age (years) | 33.7 (4.6) | 34.7 (4.71) | 32.4 (4.5) |
Gestational age (weeks) | 38.8 (0.7) | 39.02 (0.7) | 38.3 (0.9) |
%Female | 55.8 | 33.3 | 33.3 |
Avg (SEM).
p ≤ 0.0001.
METHOD DETAILS
Sample collection and processing
Placental biopsies were taken from all four quadrants of the placenta and near the umbilical cord. Decidua basalis was separated from chorionic villi immediately following delivery by experienced personnel from the dedicated maternal-fetal medicine research team at Oregon Health & Science University, Portland OR, who undergo specialized placenta collection training. Separated tissue was immersed in RPMI supplemented with 10% FBS and antibiotics then shipped overnight for processing by the Messaoudi lab. Tissues were washed in HBSS to remove contaminating blood and any visible blood vessels macroscopically separated and then washed for 10 min in HBSS before processing. Tissues were minced into approximately 0.2–0.3 mm3 cubes and enzymatically digested in 0.5 mg/mL collagenase V (Sigma, St. Louis, MO, C-9722) solution in 50 mL R3 media (RPMI 1640 with 3% FBS, 1% Penicillin-Streptomycin, 1% L-glutamine, and 1M HEPES) at 37°C for 1 h. The disaggregated cell suspension was passed through cell strainers to eliminate visible chunks of fat/tissue. Cells were pelleted from the filtrate, passed through a 70-μm cell sieve, centrifuged, and resuspended in R3 media. Red blood cells were lysed using RBC lysis buffer (155 mM NH4Cl, 12 mM NaHCO3, 0.1 mM EDTA in double-distilled water) and resuspended in 5 mL R3. The cell suspension was then layered on a discontinuous 60% and 40% percoll gradient and centrifuged for 60 min with the brakes off. Immune cells at the interface of 40% and 60% gradients were collected, washed in HBSS, counted, and cryopreserved for future analysis.
Blood processing
Peripheral blood collected before cesarean section was layered on a Ficoll-Paque (GE HealthCare Technologies Inc, Chicago, IL) gradient and centrifuged at 2000 rpm for 30 min without brakes. Peripheral Blood Mononuclear cells (PBMC) were resuspended in FetalPlex (Gemini BioProducts, West Sacramento, CA) with 10% DMSO (Sigma, St. Louis, MO) and stored in liquid nitrogen for future analysis. Plasma samples were stored at −80°C until they were used.
Immunophenotyping
For broad phenotyping of immune cells within term decidua, 1–2 X 106 fresh decidual leukocytes were washed with PBS and stained using the following cocktail of antibodies: CD45, CD20, CD4 (BioLegend, San Diego, CA), CD8b (Beckman Coulter, Brea, CA), CD14, HLA-DR, CD56, CD16, CD11c, CD123 (BioLegend, San Diego, CA), for 30 min in the dark at 4°C.
For T cell phenotyping, 0.5 X 106 freshly thawed decidual leukocytes were stained using the following panel of surface antibodies: CD8 (Beckman Coulter, Brea, CA), CD4, CD45RA, CCR7, CD69, CD103, CTLA4, CD25, and PD-1 (BioLegend, San Diego, CA) for 30 min at 4°C. Cells were then fixed and permeabilized for 30 min followed by intra-nuclear staining for FOXP3 for an additional 4 h using FOXP3 Fix/Perm Buffer set (BioLegend, San Diego, CA).
For identification of NK cell subsets, 0.5 X 106 freshly thawed decidual leukocytes were stained using the following panel of antibodies: CD56, CD16, CD103, CD49a, CD39, ITGB2, KIR2DL4, NKG2A, NKG2C, CD9 (BioLegend, San Diego, CA), KIR2DL1, CD160 (Novus Biologicals, Englewood, CO), KIR2DL2/3 (BD Biosciences, San Jose, CA).
For a more comprehensive analysis of macrophage phenotypes, 0.5 X 106 thawed decidual cells were stained using the following panel of antibodies CD14, HLA-DR, CD11c, CD16, CD68, CD86, CD163, CD206, CD209 (BioLegend, San Diego, CA), and TLR4 (BD Biosciences, San Jose, CA). A second aliquot of cells was surface stained with CD14, HLA-DR, CD64, CD9, FOLR2, CCR2 (BioLegend, San Diego, CA), TREM1, TREM2, and GLUT1 (R&D Systems, Minneapolis, MN), followed by fixation/permeabilization before staining intracellularly with TREM2 (R&D Systems, Minneapolis, MN) and S100A9 (ThermoFisher Scientific, Waltham, MA) for 4 h. Unstained and Fluorescence minus one (FMO) controls were included for all major macrophage markers.
All surface staining was done in the presence of Human Fc block and Monocyte Blocker (BioLegend, San Diego, CA). Following staining, all samples were washed twice in FACS buffer and acquired with the Attune NxT Flow Cytometer (ThermoFisher Scientific, Waltham, MA), immediately after the addition of SYTOX Live Dead Cell Stain (ThermoFisher Scientific, Waltham MA) for unfixed samples. Data were analyzed using FlowJo 10.5 (Beckton Dickinson, Ashland, OR).
Ex vivo stimulation and intracellular cytokine staining
To measure cytokine responses of decidua macrophages, 106 thawed cells were stimulated for 6h at 37°C in RPMI supplemented with 10% FBS in the presence or absence of bacterial TLR cocktail containing 1 μg/mL LPS (TLR4 ligand, E.coli 055:B5; Invivogen, San Diego, CA), 2 μg/mL Pam3CSK4 (TLR1/2 agonist, Invivogen), and 1 μg/mL FSL-1 (TLR2/6 agonist, Sigma, St. Louis, MO), or 6 X 105 cfu/mL E. coli (Escherichia coli (Migula) Castellani and Chalmers ATCC 11775).
For NK and T cell responses, 106 thawed cells were stimulated for 4h at 37°C in RPMI supplemented with 10% FBS in the presence or absence of 500 ng PMA and 1 μg Ionomycin (Sigma, St. Louis, MO). For assessment of NK cell degranulation, CD107a (BD Biosciences, San Jose, CA) was added during stimulation.
For all samples, Brefeldin A (Sigma, St. Louis, MO) was added after 1-h incubation and cell were cultured for an additional 5 h before surface and intracellular cytokine staining.
Cells were stained with the following surface antibodies: [1] CD14 and HLA-DR for macrophages; [2] CD4 and CD8 for T-cells; and [3] CD56, CD16, CD103, CD49a, CD39, and ITGB2 (BioLegend, San Diego, CA) for NK cells for 30 min in the dark at 4°C. Samples were fixed and permeabilized using fixation and permeabilization wash buffer (BioLegend, San Diego, CA) at 4°C for 20 min and stained intracellularly overnight for TNFα and IL-6, and IL-1β for macrophages, IFNγ, TNFα, IL-2, IL-4, IL-17, TGF-β and MIP-1β for T-cells and IFNγ, GZMB, and MIP-1β for NK cells. Cells were then washed and acquired using the Attune NxT Flow Cytometer (ThermoFisher Scientific, Waltham MA) and analyzed using FlowJo 10.5 (BD, Ashland OR).
Imaging flow cytometry
For visualization of macrophage subsets and nuclear translocation of NF-κB p50, decidual leukocytes were thawed, then 500,000 cells were washed and stained for dead cell exclusion (Ghost Violet 510, 1:1000 dilution) for 30 min at 4°C. Cells were then surface stained (CD14, CD11c, HLA-DR) and fixed with 1X fixation buffer (Luminex Corporation, Austin, TX) for 10 min at room temperature (RT) before the addition of p50-AF488 (1:20 dilution) in 1X Assay Buffer for 30 min at RT in the dark. At the end of the incubation, cells were washed twice in 1X Assay buffer and resuspended in 50 μL 0.25X Fixation Buffer in polypropylene Eppendorf tubes. After the addition of nuclear dye (7-AAD, 1:50), samples were run on Amnis ImageStream XMark II Imaging flow cytometer (Luminex Corporation, Austin, TX) and data were analyzed on Ideas Analysis Software (Luminex Corporation, Austin, TX).
Phospho flow
Decidual leukocytes were thawed, then washed with FACS buffer and surface stained for CD14 and HLA-DR in FACS tubes. Pellets were washed in FACS buffer and resuspended in 100 μL prewarmed PBS (Ca+ Mg+ free). Cells were fixed immediately by the addition of equal volumes of prewarmed Cytofix Buffer (BD Biosciences) and thorough mixing and incubating at 37C for 10 min. Cells were then centrifuged at 600g for 8 min. Supernatants were removed leaving no more than 50 μL residual volume. Cells were then permeabilized by the addition of 1 mL 1X BD PermWash Buffer I, mixed well, and incubated at RT for 30 min. Pellets were then spun, aspirated, and stained intranuclearly with antibodies against NF-κB p65 (pS529) AF647 (Clone K10–895.12.50, Cell Signaling Technology) or IκBa PE (Clone MFRDTRK, eBioscience, San Diego, CA) and Phospho-p38 MAPK-APC (Clone 4NIT4KK, eBioscience) for 1h at RT in the dark. Samples were washed twice in Permwash Buffer I, resuspended in FACS buffer and acquired on the Attune NxT flow cytometer.
BODIPY staining
Decidual leukocytes were thawed, then 500,000 cells surface stained (CD14, HLA-DR) for 20 min at 4°C, washed twice, and resuspended in 500 μL warm 1X PBS containing 1 μg/mL BODIPY 493/503 (ThermoFisher Scientific, Waltham, MA). Cells were incubated at 37°C for 10 min and acquired on the Attune NxT flow cytometer.
Phagocytosis assay
Decidual leukocytes were thawed, then 500,000 cells were incubated for 2 h at 37°C in media containing pH-sensitive pHrodo E. coli BioParticles conjugates (ThermoFisher Scientific, Waltham, MA) (1 mg/mL concentration). Pellets were washed twice, surface stained (CD14, HLA-DR), and resuspended in ice-cold FACS buffer. Samples were analyzed using flow cytometry with no pHrodo controls.
Cytosolic ROS assay
Decidual leukocytes were thawed, then 500,000 cells were incubated with 2.5 μM CellROX Deep Red (Life Technology, Carlsbad, CA) at 37°C for 30 min. For negative control, cells were incubated in serum-free media containing 200 μM antioxidant N-acetylcysteine (NAC) for 1.5 h. Both negative and positive controls were incubated with tert-butyl hydroperoxide (TBHP) for 30 min to induce oxidative stress. All samples were then surface stained (CD14, HLA-DR) and analyzed using flow cytometry.
Glucose uptake assay
Decidual leukocytes were thawed, then 500,000 cells were resuspended in glucose-free media and incubated with 60 mM NBDG (ThermoFisher, Waltham MA), a fluorescent glucose analog for 30 min at 37°C. Samples were washed in 1X PBS, surface stained (CD14, HLA-DR), and median fluorescence of NBDG recorded using flow cytometry.
MitoTracker analysis
Decidual leukocytes were thawed, then 500,000 cells were resuspended in 100μL of 1X MitoTracker Red CMXRos (ThermoFisher, Waltham MA) (1:1000 of stock) in 96 well plates, incubated at 37°C for 30 min, washed, surface stained (CD14, HLA-DR), and median fluorescence of MitoTracker recorded using flow cytometry.
Seahorse assay
Oxygen Consumption Rate (OCR) and Extracellular Acidification Rate (ECAR) were measured using Seahorse XF Cell Energy Phenotype kit on Seahorse XFp Flux Analyzer (Agilent Technologies, Santa Clara, CA) following the manufacturer’s instructions. Briefly, decidual leukocytes were thawed, then macrophages were sorted (pooled n = 3/group) ~200,000 cells were seeded on Cell-Tak (Corning, Corning, NY) coated 8-well culture plates in phenol-free RPMI media containing 2 mM L-glutamine, 10 mM L-glucose, and 1mM sodium pyruvate. Seeded plates were placed in 37°C incubator without CO2 then run on the XFp with extended basal measurements, followed by injection of a stressor cocktail of 1 μg/mL LPS for 1 h in a 37°C incubator without CO2. Plates were run on the XFp for 8 cycles of basal measurements, followed by acute injection of L-glucose (100 mM), oligomycin (ATP synthase inhibitor) carbonilcyanide p-triflouromethoxyphenylhydrazone (FCCP; a mitochondrial uncoupling agent).
Cytokine production
Decidual leukocytes and matched PBMCs were thawed. Live decidua macrophage subsets and peripheral blood monocytes (CD14+ HLA-DRhigh and HLA-DRlow, and SYTOX Blue negative) were sorted using BD FACS Aria Fusion. 50,000-sorted cells were placed in a 96-well plate overnight to measure cytokine production. Supernatants were collected and stored at −80°C until further analysis using Luminex (Luminex Corporation, Austin, TX). Briefly, immune mediators in cell supernatants were measured using Human Custom 29-plex Multi-Analyte Kit (R&D Systems, Minneapolis, MN) measuring the following analytes: CCL2 (MCP-1), CCL4 (MIP-1β), CXCL9 (MIG), CXCL11 (I-TAC), GM-CSF, IFNγ, IL-1RA, IL-4, IL-7, IL-12p70, IL-18, PD-L1, S100B, VEGF, IL-15, CCL3, CCL11, CXCL10, CXCL13, IFNβ, IL-1β, IL-2, IL-6, CXCL8 (IL-8), IL-17A, IL-23, PDGF-BB, TNFα, and IL-10. Samples were processed per manufacturer’s instructions, recorded, and analyzed on the Magpix xPONENT system version 4.2 (Luminex Corporation, Austin, TX).
FACS and 3′ single cell RNA library preparation
Freshly thawed immune cells from decidua, villi, maternal, and cord blood were stained with CD45-FITC at 4°C in 1% FBS in DPBS without calcium and magnesium. Cells were washed twice and sorted on BD FACS Aria Fusion into RPMI (supplemented with 30% FBS) following the addition of SYTOX Blue stain for dead cell exclusion. Cells were then counted in triplicates on a TC20 Automated Cell Counter (BioRad, Hercules, CA), washed, and resuspended in PBS with 0.04% BSA in a final concentration of 1200 cells/μL. Single-cell suspensions were then immediately loaded on the 10x Genomics Chromium Controller with a loading target of 17,600 cells. Libraries were generated using the V3 chemistry per the manufacturer’s instructions (10x Genomics, Pleasanton CA). Libraries were sequenced on Illumina NovaSeq with a sequencing target of 50,000 reads per cell.
3′ single cell RNA-Seq data analysis
Raw reads were aligned and quantified using the Cell Ranger Single-Cell Software Suite (version 3.0.1, 10x Genomics) against the GRCh38 human reference genome internally running the STAR aligner. Downstream processing of aligned reads was performed using Seurat (version 3.1.5). Droplets with ambient RNA (cells with fewer than 400 detected genes), potential doublets (cells with more than 4000 detected genes and dying cells (cells with more than 20% total mitochondrial gene expression) were excluded during initial QC. Data objects from the lean and obese groups were integrated using Seurat.87 Data normalization and variance stabilization were performed using SCTransform function87 using a regularized negative binomial regression, correcting for differential effects of mitochondrial and ribosomal gene expression levels and cell cycle.
We integrated scRNA-Seq dataset with those from matched maternal PBMC using Seurat’s IntegrateData function to remove potential contaminating blood cells. Decidual cells clustering with PBMC were removed from downstream analyses. To rule out any infiltrating fetal Hofbauer cells or monocytes, data were integrated with matched CD45+ cells isolated from chorionic villi and mononuclear cells from umbilical cord blood.
Dimensionality reduction was performed using RunPCA function to obtain the first 30 principal components followed by clustering using the FindClusters function in Seurat. Clusters were visualized using the UMAP algorithm as implemented by Seurat’s runUMAP function. Cell types were assigned to individual clusters using FindMarkers function with a fold change cutoff of at least 0.4 (FDR ≤ 0.05) and using a known catalog of well-characterized scRNA markers for human PBMC,88 tissue-resident lymphoid cells,9 and leukocytes in first-trimester human placentas.9
To study the evolution of the immune landscape of decidua with gestational age, we used the recently described atlas of first trimester (weeks 6–12) decidual CD45+ cells.9 Raw data were downloaded for 6 individuals (D6–10, D12) and analyzed as described above resulting in 24,849 cells. Cells were downsampled (6,116) to match the number of cells from term decidua of this study and integrated using Seurat’s IntegrateData function and markers compared using FindMarkers function.
Temporal trajectory was predicted using Monocle (version 2.8.0). Briefly, t-SNE was used for cell clustering. Monocle’s differentialGeneTest function was used to identify differential genes. Differential genes with a q-value less than 1e-10 were used to plot macrophages in a pseudotime scale. Metascape was used to identify Gene ontology (GO) terms.89
5′ GEX and scTCR library preparation
Matched PBMC and decidual leukocytes were thawed, washed, filtered, and stained with Ghost Violet 540 live-dead stain (Tonbo Biosciences, San Diego, CA) for 30 min in the dark at 4°C. Given the parallel assessment of both TCR and gene expression, we used TotalSeq-C, an antibody-based multiplexing technology as it is the only reagent compatible with 5′ gene expression analysis. Samples were washed thoroughly with a cell staining buffer (1X PBS with 0.5% BSA), and Fc blocked for 10 min (Human TruStain FcX, BioLegend, San Diego, CA), and incubated with a cocktail containing CD3 (SP34, BD Pharmingen), 0.5 μg each of oligo tagged CD4 (TotalSeq-C0072, BioLegend, San Diego, CA), CD8 (TotalSeq-C0046, BioLegend, San Diego, CA), CCR7 (TotalSeq-C0148, BioLegend, San Diego, CA), CD45RA (TotalSeq-C0063, BioLegend, San Diego, CA), CD69 (TotalSeq-C0146, BioLegend, San Diego, CA), CD103 (TotalSeq-C0145, BioLegend, San Diego, CA), PD-1 (TotalSeq-C0088, BioLegend, San Diego, CA), CD25 (TotalSeqTM-C0085, BioLegend, San Diego, CA), and a unique hashing antibody (TotalSeq-C0251, C0254, C0256, or C0260, BioLegend, San Diego, CA) for an additional 30 min at 4°C. Samples were washed four times with 1X PBS (serum and azide-free), filtered using Flowmi 1000 μL pipette strainers (SP Bel-Art, Wayne, NJ), and resuspended in 300 μL FACS buffer.
CD3+ T-cells were sorted on the BD FACS Aria Fusion into RPMI (supplemented with 30% FBS). Sorted cells were counted in triplicates on a TC20 Automated Cell Counter (BioRad, Hercules, CA), washed, and resuspended in PBS with 0.04% BSA in a final concentration of 1500 cells/μL. Single-cell suspensions were then immediately loaded on the 10x Genomics Chromium Controller with a loading target of 20,000 cells. Libraries were generated using the 5′ V2 chemistry (for gene expression) and Single Cell 5ʹ Feature Barcode Library Kit per manufacturer’s instructions (10x Genomics, Pleasanton CA). Libraries were sequenced on Illumina NovaSeq 6000 with a sequencing target of 30,000 gene expression reads and 10,000 feature barcode reads per cell.
5′ single cell RNA-Seq data analysis
For 5′ gene expression, alignments were performed using the feature and vdj option in Cell Ranger. Following alignment, hashing (HTO) and cell surface features (Antibody Capture) from feature barcoding alignments were manually updated in cellranger generated features file. Doublets were then removed in Seurat using the HTODemux function, which assigned sample identity to every cell in the matrix. Droplets with ambient RNA (cells with fewer than 400 detected genes) and dying cells (cells with more than 20% total mitochondrial gene expression) were excluded during initial QC. Data normalization and variance stabilization were performed on the integrated object using the NormalizeData and ScaleData functions in Seurat where a regularized negative binomial regression corrected for differential effects of mitochondrial and ribosomal gene expression levels. Dimensionality reduction was performed using the RunPCA function to obtain the first 30 principal components and clusters visualized using Seurat’s RunUMAP function. Cell types were assigned to individual clusters using FindMarkers function with a log2 fold change cutoff of at least 0.4 (FDR ≤ 0.05) and using a known catalog of well-characterized scRNA markers for human PBMC and decidual leukocytes. 5′ feature barcoding reads were normalized using centered logratio (CLR) transformation. Differential markers between clusters were then detected using FindMarkers function. A combination of gene expression and protein markers was used to define T cell subsets. CCR7 staining did not exhibit a significant positive peak, likely due to low signal in freshly thawed cells and hence was excluded from all downstream analyses. Only non-naïve T cell clusters (clusters 1, 4, 5, 7, 9, 10, 13, 5, 21, 23, 24, 26, 27, Figure S3C) based on relative gene expression of SELL, IL7R, and CCR7) and protein expression of CD45RA were retained for downstream analyses.
scTCR analyses
TCR reads were aligned to VDJ-GRCh38 ensembl reference using Cell Ranger 6.0 (10x Genomics) generating sequences and annotations such as gene usage, clonotype frequency, and cell-specific barcode information. Only cells with one productive alpha and one productive beta chain were retained for downstream analyses. CDR3 sequences were required to have a length between 5 and 27 amino acids, start with a C, and not contain a stop codon. Clonal assignments from cellranger were used to perform all downstream analyses using the R package immunarch. Data were first parsed through repLoad function in immunarch, and clonality was examined using repExplore function. Diversity estimates (Chao Diversity) were calculated using repDiversity function.
QUANTIFICATION AND STATISTICAL ANALYSIS
All statistical analyses were conducted in Prism 8 (GraphPad). All definitive outliers in two-way and four-way comparisons were identified using ROUT analysis (Q = 0.1%) after testing for normality using Shapiro-Wilk test (alpha = 0.05). If data were normally distributed across all groups, differences were tested using ordinary one-way ANOVA with unmatched samples. Multiple comparisons were corrected using Holm-Sidak test adjusting the family-wise significance and confidence level at 0.05. If the Gaussian assumption was not satisfied, differences were tested using the Kruskall-Wallis test (alpha = 0.05) followed by Dunn’s multiple hypothesis correction tests. Differences in normally distributed two groups were tested using an unpaired t test with Welch’s correction (assuming different standard deviations). Two group comparisons that failed normality tests were tested for differences using Mann-Whitney test. For subsets of cells within a sample, differences were tested using paired t-tests. p-values are indicated as # - p < 0.1, * - p < 0.05, ** - p < 0.01; *** - p < 0.001; **** - p < 0.0001, unless otherwise indicated.
Supplementary Material
Highlights.
Decidual immune changes include T cell expansion and macrophage/NK cell contraction
Term decidual T cells are skewed to Treg/Th17 and are clonally restricted
Two subsets of macrophages with distinct characteristics exist in term decidua
Maternal obesity alters decidual immunity and dampens antimicrobial responses
ACKNOWLEDGMENTS
We are grateful to all participants in this study. We thank the MFM Research Unit at Oregon Health and Science University for sample collection and Allen Jankeel, Michael Z. Zulu, Gouri Ajith, Isaac Cinco, and Hannah Debray at University of California, Irvine (UCI) for assistance with tissue processing. We thank Dr. Jennifer Atwood at the UCI Institute for Immunology Flow Cytometry Core for assistance with FACS sorting, imaging flow cytometry and Dr. Melanie Oakes at the UCI Genomics Research and Technology Hub (GRT Hub) for assistance with 10x library preparation and sequencing. This study was supported by grants from the National Institutes of Health 1K23HD06952 (N.E.M.), 1R01AI145910 (I.M.), R03AI11280 (I.M.), and 1R01AI142841 (I.M.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Footnotes
DECLARATION OF INTERESTS
The authors declare no competing interests.
INCLUSION AND DIVERSITY
We support inclusive, diverse, and equitable conduct of research.
SUPPLEMENTAL INFORMATION
Supplemental information can be found online at https://doi.org/10.1016/j.celrep.2023.112769.
REFERENCES
- 1.Maltepe E, and Fisher SJ. (2015). Placenta: the forgotten organ. Annu. Rev. Cell Dev. Biol. 31, 523–552. 10.1146/annurev-cell-bio-100814-125620. [DOI] [PubMed] [Google Scholar]
- 2.Ander SE, Diamond MS, and Coyne CB. (2019). Immune responses at the maternal-fetal interface. Sci. Immunol. 4, eaat6114. 10.1126/sciimmunol.aat6114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cappelletti M, Della Bella S, Ferrazzi E, Mavilio D, and Divanovic S. (2016). Inflammation and preterm birth. J. Leukoc. Biol. 99, 67–78. 10.1189/jlb.3MR0615-272RR. [DOI] [PubMed] [Google Scholar]
- 4.Bouç as AP, de Souza BM, Bauer AC, and Crispim D. (2017). Role of Innate Immunity in Preeclampsia: A Systematic Review. Reprod. Sci. 24, 1362–1370. 10.1177/1933719117691144. [DOI] [PubMed] [Google Scholar]
- 5.Faas MM, and De Vos P. (2018). Innate immune cells in the placental bed in healthy pregnancy and preeclampsia. Placenta 69, 125–133. 10.1016/j.placenta.2018.04.012. [DOI] [PubMed] [Google Scholar]
- 6.Geldenhuys J, Rossouw TM, Lombaard HA, Ehlers MM, and Kock MM. (2018). Disruption in the Regulation of Immune Responses in the Placental Subtype of Preeclampsia. Front. Immunol. 9, 1659. 10.3389/fimmu.2018.01659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ben Amara A, Gorvel L, Baulan K, Derain-Court J, Buffat C, Vérollet C, Textoris J, Ghigo E, Bretelle F, Maridonneau-Parini I, and Mege JL. (2013). Placental macrophages are impaired in chorioamnionitis, an infectious pathology of the placenta. J. Immunol. 191, 5501–5514. 10.4049/jimmunol.1300988. [DOI] [PubMed] [Google Scholar]
- 8.Manaster I, and Mandelboim O. (2010). The unique properties of uterine NK cells. Am. J. Reprod. Immunol. 63, 434–444. 10.1111/j.1600-0897.2009.00794.x. [DOI] [PubMed] [Google Scholar]
- 9.Vento-Tormo R, Efremova M, Botting RA, Turco MY, Vento-Tormo M, Meyer KB, Park JE, Stephenson E, Polański K, Goncalves A, et al. (2018). Single-cell reconstruction of the early maternal-fetal interface in humans. Nature 563, 347–353. 10.1038/s41586-018-0698-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Huhn O, Ivarsson MA, Gardner L, Hollinshead M, Stinchcombe JC, Chen P, Shreeve N, Chazara O, Farrell LE, Theorell J, et al. (2020). Distinctive phenotypes and functions of innate lymphoid cells in human decidua during early pregnancy. Nat. Commun. 11, 381. 10.1038/s41467-019-14123-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gustafsson C, Mjösberg J, Matussek A, Geffers R, Matthiesen L, Berg G, Sharma S, Buer J, and Ernerudh J. (2008). Gene Expression Profiling of Human Decidual Macrophages: Evidence for Immunosuppressive Phenotype. PLoS One 3, e2078. 10.1371/journal.pone.0002078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Nagamatsu T, Barrier BF, and Schust DJ. (2011). The regulation of T-cell cytokine production by ICOS-B7H2 interactions at the human fetomaternal interface. Immunol. Cell Biol. 89, 417–425. 10.1038/icb.2010.101. [DOI] [PubMed] [Google Scholar]
- 13.Abrahams VM, Kim YM, Straszewski SL, Romero R, and Mor G. (2004). Macrophages and apoptotic cell clearance during pregnancy. Am. J. Reprod. Immunol. 51, 275–282. 10.1111/j.1600-0897.2004.00156.x. [DOI] [PubMed] [Google Scholar]
- 14.Williams PJ, Bulmer JN, Searle RF, Innes BA, and Robson SC. (2009). Altered decidual leucocyte populations in the placental bed in pre-eclampsia and foetal growth restriction: a comparison with late normal pregnancy. Reproduction 138, 177–184. 10.1530/REP-09-0007. [DOI] [PubMed] [Google Scholar]
- 15.Catalano PM, and Shankar K. (2017). Obesity and pregnancy: mechanisms of short term and long term adverse consequences for mother and child. BMJ 356, j1. 10.1136/bmj.j1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gaillard R, Steegers EAP, Duijts L, Felix JF, Hofman A, Franco OH, and Jaddoe VWV. (2014). Childhood cardiometabolic outcomes of maternal obesity during pregnancy: the Generation R Study. Hypertension 63, 683–691. 10.1161/HYPERTENSIONAHA.113.02671. [DOI] [PubMed] [Google Scholar]
- 17.Lopez-Jaramillo P, Barajas J, Rueda-Quijano SM, Lopez-Lopez C, and Felix C. (2018). Obesity and Preeclampsia: Common Pathophysiological Mechanisms. Front. Physiol. 9, 1838. 10.3389/fphys.2018.01838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Salihu HM, Lynch O, Alio AP, Kornosky JL, Clayton HB, and Mbah AK. (2009). Extreme obesity and risk of placental abruption. Hum. Reprod. 24, 438–444. 10.1093/humrep/den421. [DOI] [PubMed] [Google Scholar]
- 19.Cnattingius S, Villamor E, Johansson S, Edstedt Bonamy AK, Persson M, Wikström AK, and Granath F. (2013). Maternal obesity and risk of preterm delivery. JAMA 309, 2362–2370. 10.1001/jama.2013.6295. [DOI] [PubMed] [Google Scholar]
- 20.Tersigni C, Neri C, D’Ippolito S, Garofalo S, Martino C, Lanzone A, Scambia G, and Di Simone N. (2022). Impact of maternal obesity on the risk of preterm delivery: insights into pathogenic mechanisms. J. Matern. Fetal Neonatal Med. 35, 3216–3221. 10.1080/14767058.2020.1817370. [DOI] [PubMed] [Google Scholar]
- 21.Chu SY, Kim SY, Schmid CH, Dietz PM, Callaghan WM, Lau J, and Curtis KM. (2007). Maternal obesity and risk of cesarean delivery: a meta-analysis. Obes. Rev. 8, 385–394. 10.1111/j.1467-789X.2007.00397.x. [DOI] [PubMed] [Google Scholar]
- 22.Sureshchandra S, Marshall NE, and Messaoudi I. (2019). Impact of pregravid obesity on maternal and fetal immunity: Fertile grounds for reprogramming. J. Leukoc. Biol. 106, 1035–1050. 10.1002/jlb.3ri0619-181r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Roberts KA, Riley SC, Reynolds RM, Barr S, Evans M, Statham A, Hor K, Jabbour HN, Norman JE, and Denison FC. (2011). Placental structure and inflammation in pregnancies associated with obesity. Placenta 32, 247–254. 10.1016/j.placenta.2010.12.023. [DOI] [PubMed] [Google Scholar]
- 24.Myatt L, and Maloyan A. (2016). Obesity and Placental Function. Semin. Reprod. Med. 34, 42–49. 10.1055/s-0035-1570027. [DOI] [PubMed] [Google Scholar]
- 25.Malti N, Merzouk H, Merzouk SA, Loukidi B, Karaouzene N, Malti A, and Narce M. (2014). Oxidative stress and maternal obesity: fetoplacental unit interaction. Placenta 35, 411–416. 10.1016/j.placenta.2014.03.010. [DOI] [PubMed] [Google Scholar]
- 26.Aye ILMH, Lager S, Ramirez VI, Gaccioli F, Dudley DJ, Jansson T, and Powell TL. (2014). Increasing maternal body mass index is associated with systemic inflammation in the mother and the activation of distinct placental inflammatory pathways. Biol. Reprod. 90, 129. 10.1095/biolreprod.113.116186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.St-Germain LE, Castellana B, Baltayeva J, and Beristain AG. (2020). Maternal Obesity and the Uterine Immune Cell Landscape: The Shaping Role of Inflammation. Int. J. Mol. Sci. 21, 3776. 10.3390/ijms21113776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Li Y, Chen J, Lin Y, Xu L, Sang Y, Li D, and Du M. (2021). Obesity Challenge Drives Distinct Maternal Immune Response Changes in Normal Pregnant and Abortion-Prone Mouse Models. Front. Immunol. 12, 694077. 10.3389/fimmu.2021.694077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Castellana B, Perdu S, Kim Y, Chan K, Atif J, Marziali M, and Beristain AG. (2018). Maternal obesity alters uterine NK activity through a functional KIR2DL1/S1 imbalance. Immunol. Cell Biol. 96, 805–819. 10.1111/imcb.12041. [DOI] [PubMed] [Google Scholar]
- 30.Challier JC, Basu S, Bintein T, Minium J, Hotmire K, Catalano PM, and Hauguel-De Mouzon S. (2008). Obesity in Pregnancy Stimulates Macrophage Accumulation and Inflammation in the Placenta. Placenta 29, 274–281. 10.1016/j.placenta.2007.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Frias AE, Morgan TK, Evans AE, Rasanen J, Oh KY, Thornburg KL, and Grove KL. (2011). Maternal high-fat diet disturbs uteroplacental hemodynamics and increases the frequency of stillbirth in a nonhuman primate model of excess nutrition. Endocrinology 152, 2456–2464. 10.1210/en.2010-1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yang X, Li M, Haghiac M, Catalano PM, O’Tierney-Ginn P, and Hauguel-de Mouzon S. (2016). Causal relationship between obesity-related traits and TLR4-driven responses at the maternal-fetal interface. Diabetologia 59, 2459–2466. 10.1007/s00125-016-4073-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, et al. (2014). Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 40, 274–288. 10.1016/j.immuni.2014.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Creanga AA, Catalano PM, and Bateman BT. (2022). Obesity in Pregnancy. N. Engl. J. Med. 387, 248–259. 10.1056/nejmra1801040. [DOI] [PubMed] [Google Scholar]
- 35.Catalano PM. (2007). Increasing maternal obesity and weight gain during pregnancy: the obstetric problems of plentitude. Obstet. Gynecol. 110, 743–744. 10.1097/01.AOG.0000284990.84982.ba. [DOI] [PubMed] [Google Scholar]
- 36.Sureshchandra S, Wilson RM, Rais M, Marshall NE, Purnell JQ, Thornburg KL, and Messaoudi I. (2017). Maternal Pregravid Obesity Remodels the DNA Methylation Landscape of Cord Blood Monocytes Disrupting Their Inflammatory Program. J. Immunol. 199, 2729–2744. 10.4049/jimmunol.1700434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sureshchandra S, Marshall NE, Wilson RM, Barr T, Rais M, Purnell JQ, Thornburg KL, and Messaoudi I. (2018). Inflammatory Determinants of Pregravid Obesity in Placenta and Peripheral Blood. Front. Physiol. 9, 1089. 10.3389/fphys.2018.01089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sureshchandra S, Doratt BM, Mendza N, Varlamov O, Rincon M, Marshall NE, and Messaoudi I. (2023). Maternal obesity blunts antimicrobial responses in fetal monocytes. Elife 12, e81320. 10.7554/eLife.81320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sureshchandra S, Doratt BM, Mendoza N, Varlamov O, Rincon M, Marshall NE, and Messaoudi I. (2022). Impact of pregravid obesity on anti-microbial fetal monocyte response. Preprint at bioRxiv. 10.1101/2022.07.10.499492. [DOI] [Google Scholar]
- 40.Rastogi S, Rojas M, Rastogi D, and Haberman S. (2015). Neonatal morbidities among full-term infants born to obese mothers. J. Matern. Fetal Neonatal Med. 28, 829–835. 10.3109/14767058.2014.935324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.O’Reilly JR, and Reynolds RM. (2013). The risk of maternal obesity to the long-term health of the offspring. Clin. Endocrinol. 78, 9–16. 10.1111/cen.12055. [DOI] [PubMed] [Google Scholar]
- 42.Soulas C, Conerly C, Kim WK, Burdo TH, Alvarez X, Lackner AA, and Williams KC. (2011). Recently infiltrating MAC387(+) monocytes/macrophages a third macrophage population involved in SIV and HIV encephalitic lesion formation. Am. J. Pathol. 178, 2121–2135. 10.1016/j.ajpath.2011.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yang F, Zheng Q, and Jin L. (2019). Dynamic Function and Composition Changes of Immune Cells During Normal and Pathological Pregnancy at the Maternal-Fetal Interface. Front. Immunol. 10, 2317. 10.3389/fimmu.2019.02317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.van der Zwan A, van Unen V, Beyrend G, Laban S, van der Keur C, Kapsenberg HJM, Hö llt T, Chuva de Sousa Lopes SM, van der Hoorn MLP, Koning F, et al. (2020). Visualizing Dynamic Changes at the Maternal-Fetal Interface Throughout Human Pregnancy by Mass Cytometry. Front. Immunol. 11, 571300. 10.3389/fimmu.2020.571300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Vazquez J, Chavarria M, Li Y, Lopez GE, and Stanic AK. (2018). Computational flow cytometry analysis reveals a unique immune signature of the human maternal-fetal interface. Am. J. Reprod. Immunol. 79, e12774. 10.1111/aji.12774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhang S, Ding J, Zhang Y, Liu S, Yang J, and Yin T. (2022). Regulation and Function of Chemokines at the Maternal-Fetal Interface. Front. Cell Dev. Biol. 10, 826053. 10.3389/fcell.2022.826053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Megli CJ, and Coyne CB. (2022). Infections at the maternal-fetal interface: an overview of pathogenesis and defence. Nat. Rev. Microbiol. 20, 67–82. 10.1038/s41579-021-00610-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Suryawanshi H, Morozov P, Straus A, Sahasrabudhe N, Max KEA, Garzia A, Kustagi M, Tuschl T, and Williams Z. (2018). A single-cell survey of the human first-trimester placenta and decidua. Sci. Adv. 4, eaau4788. 10.1126/sciadv.aau4788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Pavličev M, Wagner GP, Chavan AR, Owens K, Maziarz J, Dunn-Fletcher C, Kallapur SG, Muglia L, and Jones H. (2017). Single-cell transcriptomics of the human placenta: inferring the cell communication network of the maternal-fetal interface. Genome Res. 27, 349–361. 10.1101/gr.207597.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tsang JCH, Vong JSL, Ji L, Poon LCY, Jiang P, Lui KO, Ni YB, To KF, Cheng YKY, Chiu RWK, and Lo YMD. (2017). Integrative single-cell and cell-free plasma RNA transcriptomics elucidates placental cellular dynamics. Proc. Natl. Acad. Sci. USA 114, E7786–E7795. 10.1073/pnas.1710470114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Pique-Regi R, Romero R, Tarca AL, Sendler ED, Xu Y, Garcia-Flores V, Leng Y, Luca F, Hassan SS, and Gomez-Lopez N. (2019). Single cell transcriptional signatures of the human placenta in term and preterm parturition. Elife 8, e52004. 10.7554/eL-ife.52004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liu Y, Gao S, Zhao Y, Wang H, Pan Q, and Shao Q. (2021). Decidual Natural Killer Cells: A Good Nanny at the Maternal-Fetal Interface During Early Pregnancy. Front. Immunol. 12, 663660. 10.3389/fimmu.2021.663660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jabrane-Ferrat N. (2019). Features of Human Decidual NK Cells in Healthy Pregnancy and During Viral Infection. Front. Immunol. 10, 1397. 10.3389/fimmu.2019.01397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Santoni A, Carlino C, Stabile H, and Gismondi A. (2008). Mechanisms underlying recruitment and accumulation of decidual NK cells in uterus during pregnancy. Am. J. Reprod. Immunol. 59, 417–424. 10.1111/j.1600-0897.2008.00598.x. [DOI] [PubMed] [Google Scholar]
- 55.Carlino C, Stabile H, Morrone S, Bulla R, Soriani A, Agostinis C, Bossi F, Mocci C, Sarazani F, Tedesco F, et al. (2008). Recruitment of circulating NK cells through decidual tissues: a possible mechanism controlling NK cell accumulation in the uterus during early pregnancy. Blood 111, 3108–3115. 10.1182/blood-2007-08-105965. [DOI] [PubMed] [Google Scholar]
- 56.Saito S, Nishikawa K, Morii T, Enomoto M, Narita N, Motoyoshi K, and Ichijo M. (1993). Cytokine production by CD16-CD56bright natural killer cells in the human early pregnancy decidua. Int. Immunol. 5, 559–563. 10.1093/intimm/5.5.559. [DOI] [PubMed] [Google Scholar]
- 57.Ashkar AA, Di Santo JP, and Croy BA. (2000). Interferon gamma contributes to initiation of uterine vascular modification, decidual integrity, and uterine natural killer cell maturation during normal murine pregnancy. J. Exp. Med. 192, 259–270. 10.1084/jem.192.2.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Fu B, Zhou Y, Ni X, Tong X, Xu X, Dong Z, Sun R, Tian Z, and Wei H. (2017). Natural Killer Cells Promote Fetal Development through the Secretion of Growth-Promoting Factors. Immunity 47, 1100–1113.e6. 10.1016/j.immuni.2017.11.018. [DOI] [PubMed] [Google Scholar]
- 59.Whettlock EM, Woon EV, Cuff AO, Browne B, Johnson MR, and Male V. (2022). Dynamic Changes in Uterine NK Cell Subset Frequency and Function Over the Menstrual Cycle and Pregnancy. Front. Immunol. 13, 880438. 10.3389/fimmu.2022.880438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Williams PJ, Searle RF, Robson SC, Innes BA, and Bulmer JN. (2009). Decidual leucocyte populations in early to late gestation normal human pregnancy. J. Reprod. Immunol. 82, 24–31. 10.1016/j.jri.2009.08.001. [DOI] [PubMed] [Google Scholar]
- 61.de Mendonça Vieira R, Meagher A, Crespo ÂC, Kshirsagar SK, Iyer V, Norwitz ER, Strominger JL, and Tilburgs T. (2020). Human Term Pregnancy Decidual NK Cells Generate Distinct Cytotoxic Responses. J. Immunol. 204, 3149–3159. 10.4049/jimmunol.1901435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gaynor LM, and Colucci F. (2017). Uterine Natural Killer Cells: Functional Distinctions and Influence on Pregnancy in Humans and Mice. Front. Immunol. 8, 467. 10.3389/fimmu.2017.00467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Krop J, van der Zwan A, Ijsselsteijn ME, Kapsenberg H, Luk SJ, Hendriks SH, van der Keur C, Verleng LJ, Somarakis A, van der Meeren L, et al. (2022). Imaging mass cytometry reveals the prominent role of myeloid cells at the maternal-fetal interface. iScience 25, 104648. 10.1016/j.isci.2022.104648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Huang Y, Zhu XY, Du MR, and Li DJ. (2008). Human trophoblasts recruited T lymphocytes and monocytes into decidua by secretion of chemokine CXCL16 and interaction with CXCR6 in the first-trimester pregnancy. J. Immunol. 180, 2367–2375. 10.4049/jimmunol.180.4.2367. [DOI] [PubMed] [Google Scholar]
- 65.Liu L, Huang X, Xu C, Chen C, Zhao W, Li D, Li L, Wang L, and Du M. (2020). Decidual CD8+T cells exhibit both residency and tolerance signatures modulated by decidual stromal cells. J. Transl. Med. 18, 221. 10.1186/s12967-020-02371-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Sureshchandra S, Zulu MZ, Doratt B, Jankeel A, Tifrea D, Edwards R, Rincon M, Marshall NE, and Messaoudi I. (2021). Deep immune profiling of the maternal-fetal interface with mild SARS-CoV-2 infection. Preprint at bioRxiv. 10.1101/2021.08.23.457408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Sureshchandra S, Zulu MZ, Doratt BM, Jankeel A, Tifrea D, Edwards R, Rincon M, Marshall NE, and Messaoudi I. (2022). Single-cell RNA sequencing reveals immunological rewiring at the maternal-fetal interface following asymptomatic/mild SARS-CoV-2 infection. Cell Rep. 39, 110938. 10.1016/j.celrep.2022.110938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM, Prins F, van Lith JM, van der Mast BJ, Roelen DL, et al. (2010). Human decidual tissue contains differentiated CD8+ effector-memory T cells with unique properties. J. Immunol. 185, 4470– 4477. 10.4049/jimmunol.0903597. [DOI] [PubMed] [Google Scholar]
- 69.Wang SC, Li YH, Piao HL, Hong XW, Zhang D, Xu YY, Tao Y, Wang Y, Yuan MM, Li DJ, and Du MR. (2015). PD-1 and Tim-3 pathways are associated with regulatory CD8+ T-cell function in decidua and maintenance of normal pregnancy. Cell Death Dis. 6, e1738. 10.1038/cddis.2015.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Raghupathy R, Makhseed M, Azizieh F, Hassan N, Al-Azemi M, and Al-Shamali E. (1999). Maternal Th1- and Th2-type reactivity to placental antigens in normal human pregnancy and unexplained recurrent spontaneous abortions. Cell. Immunol. 196, 122–130. 10.1006/cimm.1999.1532. [DOI] [PubMed] [Google Scholar]
- 71.Taylor EB, and Sasser JM. (2017). Natural killer cells and T lymphocytes in pregnancy and pre-eclampsia. Clin. Sci. (Lond.) 131, 2911–2917. 10.1042/CS20171070. [DOI] [PubMed] [Google Scholar]
- 72.Sasaki Y, Sakai M, Miyazaki S, Higuma S, Shiozaki A, and Saito S. (2004). Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol. Hum. Reprod. 10, 347–353. 10.1093/molehr/gah044. [DOI] [PubMed] [Google Scholar]
- 73.Parker EL, Silverstein RB, Verma S, and Mysorekar IU. (2020). Viral-Immune Cell Interactions at the Maternal-Fetal Interface in Human Pregnancy. Front. Immunol. 11, 522047. 10.3389/fimmu.2020.522047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.van Egmond A, van der Keur C, Swings GMJS, Scherjon SA, and Claas FHJ. (2016). The possible role of virus-specific CD8(+) memory T cells in decidual tissue. J. Reprod. Immunol. 113, 1–8. 10.1016/j.jri.2015.09.073. [DOI] [PubMed] [Google Scholar]
- 75.Jiang X, Du MR, Li M, and Wang H. (2018). Three macrophage subsets are identified in the uterus during early human pregnancy. Cell. Mol. Immunol. 15, 1027–1037. 10.1038/s41423-018-0008-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Houser BL, Tilburgs T, Hill J, Nicotra ML, and Strominger JL. (2011). Two unique human decidual macrophage populations. J. Immunol. 186, 2633–2642. 10.4049/jimmunol.1003153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Davies LC, Jenkins SJ, Allen JE, and Taylor PR. (2013). Tissue-resident macrophages. Nat. Immunol. 14, 986–995. 10.1038/ni.2705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gomez-Lopez N, StLouis D, Lehr MA, Sanchez-Rodriguez EN, and Arenas-Hernandez M. (2014). Immune cells in term and preterm labor. Cell. Mol. Immunol. 11, 571–581. 10.1038/cmi.2014.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Aghaeepour N, Ganio EA, McIlwain D, Tsai AS, Tingle M, Van Gassen S, Gaudilliere DK, Baca Q, McNeil L, Okada R, et al. (2017). An immune clock of human pregnancy. Sci. Immunol. 2, eaan2946. 10.1126/sciimmunol.aan2946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sureshchandra S, Marshall NE, Mendoza N, Jankeel A, Zulu MZ, and Messaoudi I. (2021). Functional and genomic adaptations of blood monocytes to pregravid obesity during pregnancy. iScience 24, 102690. 10.1016/j.isci.2021.102690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.True H, Blanton M, Sureshchandra S, and Messaoudi I. (2022). Monocytes and macrophages in pregnancy: The good, the bad, and the ugly. Immunol. Rev. 308, 77–92. 10.1111/imr.13080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Norman SM, Tuuli MG, Odibo AO, Caughey AB, Roehl KA, and Cahill AG. (2012). The effects of obesity on the first stage of labor. Obstet. Gynecol. 120, 130–135. 10.1097/AOG.0b013e318259589c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Frolova AI, Wang JJ, Conner SN, Tuuli MG, Macones GA, Woolfolk CL, and Cahill AG. (2018). Spontaneous Labor Onset and Outcomes in Obese Women at Term. Am. J. Perinatol. 35, 59–64. 10.1055/s-0037-1605574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Laskewitz A, van Benthem KL, Kieffer TEC, Faas MM, Verkaik-Schakel RN, Plösch T, Scherjon SA, and Prins JR. (2019). The influence of maternal obesity on macrophage subsets in the human decidua. Cell. Immunol. 336, 75–82. 10.1016/j.cellimm.2019.01.002. [DOI] [PubMed] [Google Scholar]
- 85.Lindau R, Vondra S, Spreckels J, Solders M, Svensson-Arvelund J, Berg G, Pollheimer J, Kaipe H, Jenmalm MC, and Ernerudh J. (2021). Decidual stromal cells support tolerance at the human foetalmaternal interface by inducing regulatory M2 macrophages and regulatory T-cells. J. Reprod. Immunol. 146, 103330. 10.1016/j.jri.2021.103330. [DOI] [PubMed] [Google Scholar]
- 86.Marshall NE, Biel FM, Boone-Heinonen J, Dukhovny D, Caughey AB, and Snowden JM. (2019). The Association between Maternal Height, Body Mass Index, and Perinatal Outcomes. Am. J. Perinatol. 36, 632–640. 10.1055/s-0038-1673395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, Hao Y, Stoeckius M, Smibert P, and Satija R. (2019). Comprehensive Integration of Single-Cell Data. Cell 177, 1888–1902.e21. 10.1016/j.cell.2019.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zheng GXY, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, Ziraldo SB, Wheeler TD, McDermott GP, Zhu J, et al. (2017). Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049. 10.1038/ncomms14049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, and Chanda SK. (2019). Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523. 10.1038/s41467-019-09234-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The datasets supporting the conclusions of this article are available on NCBI’s Sequence Read Archive and project ID’s are listed in the key resources table.
This paper does not report original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
KEY RESOURCES TABLE
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
| ||
Antibodies | ||
| ||
CD3-FITC | BD Biosciences | Cat#556611; RRID:AB_396484 Clone-SP34 |
CD20-BV510 | BioLegend | Cat#302340; RRID:AB_2561941 Clone-2H7 |
CD56-BV711 | BioLegend | Cat#318336; RRID:AB_2562417 Clone-HCD56 |
CD16-PB | BioLegend | Cat#302021; RRID:AB_492978 Clone-3G8 |
CD14-AF700 | BioLegend | Cat#301822; RRID:AB_493747 Clone-M5E2 |
HLA-DR-APC-Cy7 | BioLegend | Cat#307618; RRID:AB_493586 Clone-L243 |
CD11c-APC | BioLegend | Cat#301614; RRID:AB_493023 Clone-3.9 |
CD123-PCP-Cy5.5 | BioLegend | Cat#306016; RRID:AB_2264693 Clone-6H6 |
CD86-BV605 | BioLegend | Cat#305430; RRID:AB_2563824 Clone-IT2.2 |
CD4-FITC | BioLegend | Cat#300538; RRID:AB_2562052 Clone-RPA-T4 |
CD4-PE | BioLegend | Cat#317410; RRID:AB_571955 Clone-OKT4 |
CD8b-ECD | Beckman Coulter | Cat#6607123; RRID:AB_1575983 Clone-2ST8.5H7 |
CD45RA-PerCP-Cy5.5 | Thermo Fisher Scientific | Cat#45-0458-42; RRID:AB_10718536 Clone-HI100 |
CCR7-PE-Cy7 | BioLegend | Cat#353226; RRID:AB_11126145 Clone-GO43H7 |
CD69-PE | BioLegend | Cat#310906; RRID:AB_314841 Clone-FN50 |
CD103-BV605 | BioLegend | Cat#350218; RRID:AB_2564283 Clone-BER Act1 |
CTLA4-PeDazzle594 | BioLegend | Cat#369616; RRID:AB_2632878 Clone-BNI3 |
CD25-APC-Cy7 | BioLegend | Cat#302614; RRID:AB_314284 Clone-BC96 |
FOXP3-A488 | BioLegend | Cat#320106; RRID:AB_439752 Clone-206D |
CD49a-PE-Cy7 | BioLegend | Cat#328312; RRID:AB_2566272 Clone-TS2/7 |
CD39-BV510 | BioLegend | Cat#328219; RRID:AB_2563265 Clone-A1 |
ITGB2 (CD11a/CD18)-PCP-Cy5.5 | BioLegend | Cat#302120; RRID:AB_2565587 Clone-TS1/18 |
KIR2DL4 (CD158d)-APC | BioLegend | Cat#347008; RRID:AB_2130691 Clone-mAb33 |
KIR2DL1 (CD158a)-APC-Cy7 | Novus Biologicals | Cat#NB100-63267APCCY7; RRID:AB_2941073 Clone-NKVFS1 |
KIR2DL2/3 (CD158b)-PE | BD Biosciences | Cat#559785; RRID:AB_397326 Clone-CH-L |
CD160-AF700 | Novus Biologicals | Cat#NBP2-90074AF700; RRID:AB_2941072 Clone-275 |
NKG2A-PE-Dazzle594 | BioLegend | Cat#375121; RRID:AB_2888869 Clone-S19004C |
NKG2C-PE | BioLegend | Cat#375003; RRID:AB_2888871 Clone-S19005E |
CD9-PE-Cy7 | BioLegend | Cat#312116; RRID:AB_2728256 Clone-HI9a |
CD68-APC | BioLegend | Cat#333810; RRID:AB_2275735 Clone-Y1/82A |
TLR4-BV711 | BD Biosciences | Cat#564404; RRID:AB_2738794 Clone-tf901 |
CD163-APC/Fire750 | BioLegend | Cat#333634; RRID:AB_2734333 Clone-GHI/61 |
CD206-BV510 | BioLegend | Cat#321138; RRID:AB_2721530 Clone-15-2 |
CD209-FITC | BioLegend | Cat#330104; RRID:AB_1134048 Clone-9e9a8 |
TREM1-AF405 | R&D Systems | Cat#FAB12781V; RRID:AB_2941074 Clone-888111 |
TREM2-PerCP | R&D Systems | Cat#FAB17291C; RRID:AB_2941075 Clone-237920 |
CD64-BV711 | BioLegend | Cat#305042; RRID:AB_2800778 Clone-10.1 |
CD64-APC | BioLegend | Cat#305014; RRID:AB_1595428 Clone-10.1 |
GLUT1-AF488 | R&D Systems | Cat#FAB1418G; RRID:AB_2941076 Clone-202915 |
FOLR2-PE | BioLegend | Cat#391704; RRID:AB_2721336 Clone-94b/folr2 |
CCR2-BV605 | BioLegend | Cat#357213; RRID:AB_2562702 Clone-K036C2 |
CD19-PE | BioLegend | Cat#302208; RRID:AB_314238 Clone-HIB19 |
S100A9-APC | Invitrogen | Cat#MA5-28129; RRID:AB_2745112 Clone-10.1 |
CD107a-FITC | BD Biosciences | Cat#555800; RRID:AB_396134 Clone-H4A3 |
PD-1-BV510 | BioLegend | Cat#329932; RRID:AB_2562256 Clone-Eh12.2h7 |
CD45-FITC | BioLegend | Cat#304038; RRID:AB_2562050 Clone-HI30 |
TNFα-APC | BioLegend | Cat#502912; RRID:AB_315264 Clone-Mab11 |
IL-6-FITC | BD Biosciences | Cat#554696; RRID:AB_395514 Clone-MQ2-6A3 |
IL-1β-PB | BioLegend | Cat#511710; RRID:AB_2124350 Clone-H1b-98 |
IFNγ-APC | Invitrogen | Cat#17-7319-82; RRID:AB_469506 Clone-4S.B3 |
IFNγ-PE-Cy7 | eBioscience | Cat#502527; RRID:AB_1626154 Clone-4S.B3 |
IL-2-AF700 | BioLegend | Cat#500320; RRID:AB_528929 Clone-MQ-17H12 |
IL-4-APC | Tonbo Biosciences | Cat#50-210-2786; RRID:AB_2941077 Clone-MP425D2 |
IL-17-FITC | Invitrogen | Cat#11-717942; RRID:AB_10805390 Clone-Ebio64dec17 |
TGF-β-PerCP-Cy5.5 | BioLegend | Cat#141410; RRID:AB_2561592 Clone-TW7-16B4 |
MIP-1β-PE | BD Biosciences | Cat#550078; RRID:AB_393549 Clone-D21-1351 |
GZMB-AF700 | BD Biosciences | Cat#560213; RRID:AB_1645453 Clone-GB11 |
NF-κB p50-AF488 | Luminex | Cat#4700-1674; RRID:AB_2941078 |
7-Amino-Actinomycin D (7-AAD) | Luminex | Cat#400-0290; RRID:AB_2941079 |
NF-κB p65 (pS529)-AF647 | BD Biosciences | Cat#558422; RRID:AB_647136 Clone-K10-895.12.50 |
IκBa-PE | eBioscience | Cat#12-903642; RRID:AB_2572683 Clone-MFRDTRK |
Phospho-p38 MAPK-APC | eBiosceince | Cat#17-9078-42; RRID:AB_2573290 Clone-4NIT4KK |
CD3-PE | BD Biosciences | Cat#556612; RRID:AB_396485 Clone-SP34 |
TotalSeq-C0072-CD4 | BioLegend | Cat#300567; RRID:AB_2800725 Clone-RPA-T4 |
TotalSeq-C0046-CD8 | BioLegend | Cat#344753; RRID:AB_2800922 Clone-SK1 |
TotalSeq-C0148-CCR7 | BioLegend | Cat#353251; RRID:AB_2800943 Clone-G043H7 |
TotalSeq-C0063-CD45RA | BioLegend | Cat#304163; RRID:AB_2800764 Clone-HI100 |
TotalSeq-C0146-CD69 | BioLegend | Cat#310951; RRID:AB_2800810 Clone-FN50 |
TotalSeq-C0145-CD103 | BioLegend | Cat#350233; RRID:AB_2800933 Clone-Ber-ACT8 |
TotalSeq-C0088-PD-1 | BioLegend | Cat#329963; RRID:AB_2800862 Clone-EH12.2H7 |
TotalSeq-C0085-CD25 | BioLegend | Cat#302649; RRID:AB_2800745 Clone-BC96 |
TotalSeq-C0251 | BioLegend | Cat#394661; RRID:AB_2801031 Clone-LNH-94, 2M2 |
TotalSeq-C0254 | BioLegend | Cat#394667; RRID:AB_2801034 Clone-LNH-94, 2M2 |
TotalSeq-C0256 | BioLegend | Cat#394671; RRID:AB_2820042 Clone-LNH-94, 2M2 |
TotalSeq-C0260 | BioLegend | Cat#394679; RRID:AB_2820046 Clone-LNH-94, 2M2 |
| ||
Biological Samples | ||
| ||
Fetal Bovine Serum, USDA Certified, Heat Inactivated | Omega Scientific | Cat#FB-02 |
FetalPlex™ Animal Serum Complex | GeminiBioProducts | Cat#100-602 |
| ||
Chemicals, Peptides, and Recombinant Proteins | ||
| ||
RPMI 1640 | VWR | Cat#45000-396 |
Hanks Balanced Salt Solution (HBSS) | VWR | Cat#21-023-CV |
Penicillin Streptomycin | GeminiBio | Cat#400109 |
L-Glutamine | GeminiBio | Cat#400106 |
N-2-hydroxyethylpiperazine-N′2-ethanesulfonic acid HEPES | ThermoFisher | Cat#15630080 |
Collagenase | Sigma | Cat# C9722 Source: Clostridium histolyticum |
Ammonium chloride - NH4Cl (RBC Lysis Buffer) | Sigma | Cat#A9434 |
Sodium hydrogen carbonate - CHNaO3 (RBC Lysis Buffer) | Fisher | Cat#AAA170050E |
EDTA (RBC Lysis Buffer) | Invitrogen | Cat#AM9260G |
Percoll Density Gradient | Neta Scientific | Cat# 17-0891-01 |
Ficoll-Paque | GE Healthcare | Cat# 17144003 |
Dimethyl sulfoxide (DMSO) | Sigma | Cat# D2650-100ML |
FOXP3 Fix/Perm Buffer Set | BioLegend | Cat#421403 |
Ghost Dye 510 | TONBO Biosciences | Cat#13-0870-T 100 |
Ghost Dye 540 | TONBO Biosciences | Cat#13-0879 |
SYTOX Blue Dead Cell Stain | ThermoFisher Scientific | Cat#S34857 |
Human TruStain FcX | BioLegend | Cat#422302 |
Human True-Stain Monocyte Blocker | BioLegend | Cat#426103 |
Lipopolysaccharide (LPS) TLR4 ligand E. coli 055:B5 | Invivogen | Cat#tlrl-b5lps |
Pam3CSK4 TLR1/2 agonist | Invivogen | Cat#tlrl-pm2s-1 |
FSL-1 TLR2/6 agonist | Sigma | Cat#SML1420 |
E. coli | Migula. Castellani and Chalmers ATCC 11775 | Cat#1132342 |
Phorbol myristate acetate-NF-κB Activator (PMA) | Invivogen | Cat#tlrl-pma |
Ionomycin | Invivogen | Cat#inh-ion |
Brefeldin A (BFA) | BioLegend | Ca#420601 |
Fixation buffer | BioLegend | Cat#420801 |
Amnis NF-κB translocation kit | Luminex Corporation | Cat #ACS10000 |
Permeabilization wash buffer | BioLegend | Cat#421002 |
Bovine Serum Albumin (BSA), Fraction V— Molecular Biology Grade |
GeminiBio | Cat#700-106P |
Cytofix fixation buffer | BD Biosciences | Cat#554655 |
BD Perm/Wash buffer | BD Biosciences | Cat#554723 |
| ||
Critical Commercial Assays | ||
| ||
BODIPY 493/503 | ThermoFisher Scientific | Cat#D3922 |
pHrodo Deep Red E. coli BioParticle Conjugates | ThermoFisher Scientific | Cat#P35360 |
CellROX Deep Red Flow Cytometry Assay Kit | ThermoFisher Scientific | Cat#C10491 |
2-NBDG | ThermoFisher Scientific | Cat#N13195 |
MitoTracker Red CMXRos | ThermoFisher Scientific | Cat#M7512 |
Seahorse XF Cell Energy Phenotype kit | Agilent Technologies | Cat#103592-100 |
Human Premixed 29-plex Magnetic Luminex Assay | R&D Systems | Cat#Custom-LxSA-H-29 Lot#C0003514 |
TotalSeq B Hashtag Oligos 1-10 | BioLegend | 384631-392649 |
Chromium Single Cell 3’ Reagent Kits v3 | 10X Genomics | PN-1000075 |
3′ Feature Barcode Kit | 10X Genomics | PN-1000262 |
Chromium Single Cell 5’ Reagent Kits V2 | 10X Genomics | PN-1000263 |
5′ Feature Barcode Kit | 10X Genomics | PN-1000256 |
| ||
Deposited Data | ||
| ||
3′ Single Cell GEX data - Decidua | This paper | NCBI Sequence Read Archive: PRJNA817521 |
5′ Single Cell TCR data - Decidua | This paper | NCBI Sequence Read Archive: PRJNA817521 |
5′ Single Cell TCR data - Maternal PBMC | This paper | NCBI Sequence Read Archive: PRJNA817521 |
3′ Single Cell GEX data -Villous | This paper | NCBI Sequence Read Archive:PRJNA946160 |
3′ Single Cell GEX data - UCBMC | Sureshchandra et al. 202166 10.3389/fimmu.2021.617592 |
NCBI Sequence Read Archive:PRJNA690128 |
3′ Single Cell GEX data - Maternal PBMC | This paper and Sureshchandra etal. 202166 10.1016/j.isci.2021.102690 |
NCBI Sequence Read Archive:PRJNA887020 |
| ||
Software and Algorithms | ||
| ||
Prism | GraphPad | Version#8 |
Immunarch | R package | Version#0.6.7 |
Metascape | www.metascape.org | N/A |
Attune NxT Flow Cytometer Software | ThermoFisher Scientific | Version#2.5 |
FlowJo | TreeStar | Version#10.5 |
Ideas Analysis Software | Luminex Corporation | Version#6.1 |
Seahorse Wave | Agilent Technologies | Version#2.6.1 |
xPONENT® Software for Luminex Instruments | Luminex Corporation | Version#4.3 |
Cell Ranger Single-Cell Software Suite | 10X Genomics | Version 6.0.2 |
Seurat | R package | Version 3.1.5 |
Monocle | R package | Version 2.8.0 |