International Journal of Molecular Sciences

Article Cell–Cell Communication at the Embryo Implantation Site of Mouse Uterus Revealed by Single-Cell Analysis

Yi Yang 1, Jia-Peng He 2 and Ji-Long Liu 2,*

1 College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; [email protected] 2 Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; [email protected] * Correspondence: [email protected]

Abstract: As a crucial step for human reproduction, embryo implantation is a low-efficiency process. Despite rapid advances in recent years, the molecular mechanism underlying embryo implantation remains poorly understood. Here, we used the mouse as an animal model and generated a single- cell transcriptomic atlas of embryo implantation sites. By analyzing inter-implantation sites of the uterus as control, we were able to identify global expression changes associated with embryo implantation in each cell type. Additionally, we predicted signaling interactions between uterine luminal epithelial cells and mural trophectoderm of blastocysts, which represent the key mechanism of embryo implantation. We also predicted signaling interactions between uterine epithelial-stromal crosstalk at implantation sites, which are crucial for post-implantation development. Our data provide a valuable resource for deciphering the molecular mechanism underlying embryo implantation.   Keywords: embryo implantation; mouse; single-cell RNA-seq; transcriptional changes Citation: Yang, Y.; He, J.-P.; Liu, J.-L. Cell–Cell Communication at the Embryo Implantation Site of Mouse Uterus Revealed by Single-Cell 1. Introduction Analysis. Int. J. Mol. Sci. 2021, 22, Embryo implantation is a crucial step for human reproduction. This is a low-efficiency 5177. https://doi.org/10.3390/ process and the implantation failure rate per menstrual cycle is approximately 70% [1,2]. ijms22105177 In assisted reproductive technologies, despite a high success rate of in vitro fertilization, implantation rate remains very low [3]. Additionally, an emerging concept is that early Academic Editor: Chaya Kalcheim pregnancy loss and various pregnancy complications are rooted in suboptimal embryo implantation [4]. Therefore, it is imperative to understand the molecular mechanism of Received: 23 April 2021 embryo implantation. Accepted: 10 May 2021 Published: 13 May 2021 Due to ethical restrictions and experimental difficulties, in vivo analysis of embryo im- plantation heavily relies on mice. By using uterus-specific gene knockout mice, a number of

Publisher’s Note: MDPI stays neutral have been proven to be required for embryo implantation [5]. Alternatively, several with regard to jurisdictional claims in studies have analyzed global changes between implantation sites and published maps and institutional affil- inter-implantation sites of mouse uterus using high-throughput transcriptomic approaches, iations. providing a useful candidate gene list for further study of embryo implantation [6–8]. The limitation of these studies is that the whole uterus is used. The uterus has a complex struc- ture consisting of three layers, namely the endometrium, myometrium and perimetrium, and many cell types, including luminal and glandular epithelial cells, stromal cells, smooth muscle cells, endothelial cells and various immune cells. Thus, a whole uterus bulk RNA- Copyright: © 2021 by the authors. Licensee MDPI, Basel, Switzerland. seq approach is unable to accurately capture cell-type-specific gene expression changes. This article is an open access article In addition to whole uteri, isolated uterine luminal epithelium from implantation sites distributed under the terms and and inter-implantation sites by enzymes [9] and laser-capture microdissection (LCM) [10] conditions of the Creative Commons have been subjected to high-throughput transcriptomic analysis. It is undoubtedly true Attribution (CC BY) license (https:// that the use of isolated uterine luminal epithelium was a better choice than the whole creativecommons.org/licenses/by/ uterus. However, the limitation is that the contributions of cell types, other than luminal 4.0/). epithelium, and signaling crosstalk between different cell types were not considered.

Int. J. Mol. Sci. 2021, 22, 5177. https://doi.org/10.3390/ijms22105177 https://www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2021, 22, 5177 2 of 13

With advances in single-cell RNA-seq techniques, it is now possible to analyze global gene expression profiles within highly heterogeneous tissues at the single-cell level [11]. In the present study, by using the-state-of-the-art single-cell RNA-seq approach, we resolved all cell types at implantation sites and inter-implantation sites of the mouse uterus on gestational day 5. Consequently, we were able to identify different expression changes associated with embryo implantation in all cell types. Additionally, we predicted that cell– cell communication is involved in embryo implantation. Our study provides a valuable resource for understanding the molecular mechanisms underlying embryo implantation in mice.

2. Results 2.1. Single-Cell Analysis of Embryo Implantation Site in Mouse Uterus Embryo implantation takes place on gestational day 5 in mice. The implantation site in the uterus can be visualized after an intravenous injection of Chicago Blue B dye solution (Figure1A). In order to capture a cell-type resolved map of mouse embryo implantation, single-cell RNA-seq data were generated from the implantation site (IS) and the inter- implantation site (IIS, served as control) by using a 10x Genomics platform. The whole uterus, which consists of the endometrium, myometrium and perimetrium, was used. The blastocyst at IS was also kept. After quality control, a total of 8421 cells (2606 for IS and 5815 for IIS) were obtained. Unsupervised clustering analysis revealed 19 distinct cell clusters for IS and IIS combined (Figure1B). Major cell types were defined using the expression of known cell-type-specific genes, with stromal cells expressing Hoxa11 [12], epithelial cells expressing Krt8 [13], smooth muscle cells expressing Acta2 [14], pericytes expressing Rgs5 [15], endothelial cells expressing Vwf or Prox1 [16] and immune cells expressing Ptprc [17] (Figure1C). We found four stromal cell clusters, S1, S1p, S2 and S2p. Cells in S1/S1p but not S2/S2p expressed high levels of Hand2, implying that S1/S1p were inner stromal cells and S2/S2p were outer stromal cells [18]. S1p and S2p expressed high level of Mki67, suggesting that S1p and S2p were a subset of proliferating S1 and S2 cells, respec- tively. There were two epithelial cell clusters, LE and GE. LE contained luminal epithelial cells expressing Tacstd2, and GE was composed of glandular epithelial cells expressing Foxa2 [19]. Only one smooth muscle cell cluster and one pericyte cluster were identi- fied. Endothelial cells had four clusters: VEC and its proliferating subset VECp were vascular endothelial cells expressing Vwf, while LEC and its proliferating subset LECp were lymphatic endothelial cells expressing Prox1 [16]. The seven immune cell clusters were macrophages (M, Ptprc+Adgre1+)[20], a proliferating subset of macrophages (Mp, Ptprc+Adgre1+Mki67+), dendritic cells (DC, Ptprc+Cd209a+Siglech−)[20], plasmacytoid dendritic cells (pDC, Ptprc+Cd209a+Siglech+)[21], mixed natural killer cells and T cells (NK/T, Ptprc+Nkg7+ or Ptprc+Cd3e+)[22], a proliferating subset of natural killer cells (NKp, Ptprc+Nkg7+Cd3e−Mki67+) and B cells (B, Ptprc+Cd79a+)[22]. We next aimed to discover novel markers for each cell type. We selected genes that were expressed significantly higher in the cell type of interest than the other cell types by Wilcoxon rank sum test. A heatmap depicting the top 10 marker genes for each cell type is shown in Figure1D. The complete lists of marker genes are presented in Supplementary Table S1. Int. J. Mol. Sci. 2021, 22, 5177 3 of 13 Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 3 of 13

FigureFigure 1. 1.Single-cell Single-cell transcriptome transcriptome analysis analysis of of the the implantation implantation site site in in mouse mouse uterus uterus on on gestational gestational day day 5: (5:A )(A A) photographA photo- ofgraph mouse of uterus mouse on uterus gestational on gestational day 5. day The 5. position The position of embryo of embryo implantation implantation sites sites was was marked marked with with an an arrow. arrow. (B ()B The) The t-stochastic neighbor embedding (tSNE) representation of single-cell RNA-seq data obtained from implantation sites t-stochastic neighbor embedding (tSNE) representation of single-cell RNA-seq data obtained from implantation sites (IS) and (IS) and inter-implantation sites (IIS). Single cells were grouped by cellular origin (left) and cell clusters (right). LE: lu- inter-implantationminal epithelial cells; sites (IIS).GE: glandular Single cells epithelial were grouped cells; S1: by superf cellularicial origin stromal (left) cells; and S2: cell deep clusters stromal (right). cells; LE: S1p: luminal proliferati epithelialng cells;superficial GE: glandular stromal epithelial cells; S2p: cells; proliferating S1: superficial deep stromalstromal cells;cells; S2:SMC: deep smooth stromal muscle cells; S1p:cells; proliferatingPC: pericytes; superficial VEC: vascular stromal cells;endothelial S2p: proliferating cells; VECp: deep proliferating stromal cells; vascular SMC: smoothendothelial muscle cells; cells; LEC: PC: lymphatic pericytes; endothelia VEC: vascularl cells; endothelial VECp: proliferating cells; VECp: proliferatinglymphatic vascularendothelial endothelial cells; M: cells;macrophages; LEC: lymphatic DC: dendritic endothelial cells; cells; pDC: VECp: plasmacytoid proliferating dendritic lymphatic cells; Mp: endothelial proliferating cells; M: macrophages;macrophages; DC: NK/T: dendritic mixed cells; natural pDC: killer plasmacytoid cells and T dendritic cells; NKp: cells; proliferating Mp: proliferating natural macrophages;killer cells; B: NK/T:B cells. mixed(C) The natural ex- pression pattern of canonical marker genes projected onto TSNE plots. Dashed lines denote the boundaries of the cell killer cells and T cells; NKp: proliferating natural killer cells; B: B cells. (C) The expression pattern of canonical marker cluster of interest. (D) Heatmap of the top 10 gene expression signatures for each cell type. genes projected onto TSNE plots. Dashed lines denote the boundaries of the cell cluster of interest. (D) Heatmap of the top 10 gene expression signatures for each cell type. Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 4 of 13 Int. J. Mol. Sci. 2021, 22, 5177 4 of 13

2.2. Cell Type Proportion Changes at Implantation Site Compared to Inter-Implantation Site 2.2. Cell Type Proportion Changes at Implantation Site Compared to Inter-Implantation Site WeWe investigated investigated the the abundance abundance of eacheach cellcell typetype at at IS IS compared compared to to IIS. IIS. For For each each cell cell 2 type,type, proliferating proliferating and and non-proliferating non-proliferating cells cells were were summed. summed. The Theχ2 χtest test was was employed employed to to assessassess the the significance significance of of difference difference betweenbetween twotwo groups. groups. By By using using the the criteria criteria of pof< p 0.05 < 0.05 andand fold fold change change >2, > 2, the the proportions proportions of of all cell typestypes remainedremained unchanged, unchanged, except except for for LEC LEC andand B Bcells cells (Figure (Figure 22A).A). Of Of special special interest, interest, we we investigated investigated the abundancethe abundance of proliferating of proliferat- ingcells. cells. We We found found that that the the proportions proportions of Mp of Mp and and NKp NKp were were unchanged unchanged in IS in compared IS compared to toIIS. IIS. Notably, Notably, the the proportion proportion of LECpof LECp significantly significantly decreased, decreased, whereas whereas the proportions the proportions of ofS1p, S1p, S2p S2p and and VECp VECp significantly significantly increased increased (Figure (Figure2B). 2B).

FigureFigure 2. Cell 2. Cell population population shifts shifts in implantation in implantation sites sites compared compared to inter-implantation to inter-implantation sites: sites: (A) (PieA) Pieplots plots showing showing the thecell cellpopu- lationpopulation change of change 13 major of 13 cell major types cell in typesimplantation in implantation sites (IS) sites compared (IS) compared to inter-implantation to inter-implantation sites (IIS) sites on (IIS)gestational on gestational day 5. For eachday cell 5. type, For eachproliferating cell type, and proliferating non-proliferating and non-proliferating cells were summed; cells were(B) Bar summed; plot showing (B) Bar cell plot population showing cellchanges population of prolif- 2 eratingchanges cells. of Cell proliferating types with cells. fold Cell change types (based with foldon percentage) change (based > 2 onand percentage) p-value (χ > test) 2 and < 0.05p-value were (χ labeled2 test) < in 0.05 red. were labeled in red. 2.3. Cell-Type-Specific Transcriptional Changes Associated with Embryo Implantation 2.3.We Cell-Type-Specific investigated Transcriptionalthe breadth of Changes transcriptional Associated withchanges Embryo in Implantationeach cell type by per- formingWe differential investigated gene the breadth expression of transcriptional analysis (Figure changes 3A). in eachUsing cell a typelogFC by performingcutoff of 0.25 anddifferential a p-value gene cutoff expression of 0.05, we analysis identified (Figure 391,3A). 176, Using 281, a 777, logFC 385, cutoff 369, of 305, 0.25 199, and 211, a p -value592, 422, 513,cutoff 95, of84, 0.05, 310, we426, identified 323, 335 391,and 176,309 differentially 281, 777, 385, expressed 369, 305, 199, genes 211, for 592, LE, 422, GE, 513, S1, 95, S1p, 84, S2, S2p,310, SMC, 426, 323,PC, 335VEC, and VECp, 309 differentially LEC, LECp, expressed NK/T, NK genesp, B, forM, LE,Mp, GE, DC S1, and S1p, pDC, S2, S2p,respectively SMC, (FigurePC, VEC, 3B VECp,and Supplementary LEC, LECp, NK/T, Table NKp, S2). B,We M, th Mp,en explored DC and pDC, the biological respectively implications (Figure3B of and Supplementary Table S2). We then explored the biological implications of differentially differentially expressed genes using (GO) analysis. A complete list of en- expressed genes using gene ontology (GO) analysis. A complete list of enriched GO terms riched GO terms is provided in Figure 3C. These data indicated that each cell type invokes is provided in Figure3C. These data indicated that each cell type invokes distinct biological distinctprocesses biological in order processes to participate in order in embryo to participate implantation. in embryo implantation. Int. J. Mol. Sci. 2021, 22, 5177 5 of 13 Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 5 of 13

Figure 3. Gene expression changes in implantation sitessites compared to inter-implantationinter-implantation sites: (A) Heatmap showing the distribution of differentially differentially ex expressedpressed genes genes (logFC (logFC > >0.25 0.25 and and p p< <0.05) 0.05) between between implantation implantation sites sites (IS) (IS) compared compared to in- to ter-implantation sites (IIS) in each cell type; (B) the count of differentially expressed genes in each cell type; (C) gene on- inter-implantation sites (IIS) in each cell type; (B) the count of differentially expressed genes in each cell type; (C) gene tology (GO) enrichment analysis of downregulated genes and upregulated genes, respectively. p < 0.05 was used as the ontology (GO) enrichment analysis of downregulated genes and upregulated genes, respectively. p < 0.05 was used as the significance cutoff. Non-significant hits (p ≥ 0.05) are depicted in gray. significance cutoff. Non-significant hits (p ≥ 0.05) are depicted in gray. 2.4. Inferring Cell–Cell Communication between Blastocyst and Uterus at the Implantation Site 2.4. Inferring Cell–Cell Communication between Blastocyst and Uterus at the Implantation Site The cell–cell communication between blastocyst and uterus represents the key mechanismThe cell–cell of embryo communication implantation. between Due blastocystto the relatively and uterus small represents number theof keyblastocyst mech- cellsanism at ofthe embryo embryo implantation. implantation Due site, towe the did relatively not find smallany blastocyst number of-related blastocyst cell clusters cells at inthe our embryo single-cell implantation RNA-seq site, data we (data did not findshown). any Alternatively, blastocyst-related we re-analyzed cell clusters a in pub- our lishedsingle-cell single-cell RNA-seq RNA-seq data (data dataset not on shown). mouse Alternatively, E4.5 blastocysts we re-analyzed[23] (Figure a4A). published E4.5 is equivalentsingle-cell RNA-seqto gestational dataset day on 5 in mouse our study. E4.5 blastocysts By using canonical [23] (Figure marker4A). E4.5genes, is equivalentfour major cellto gestational types were day identified: 5 in our study.epiblast By (EPI) using (F canonicaligure 4B), marker primitive genes, endoderm four major (PE) cell (Figure types 4C),were polar identified: trophectoderm epiblast (EPI)(pTE) (Figureand mural4B), trophectoderm primitive endoderm (mTE) (PE)(Figure (Figure 4D).4 C), polar trophectoderm (pTE) and mural trophectoderm (mTE) (Figure4D). During mouse embryo implantation, the blastocysts attach to the LE of the uterus in the way of mTE. Thus, we used CellPhoneDB (https://www.cellphonedb.org/ (ac- cessed 22 April 2021)) to predict the ligand–receptor interactions between LE and mTE. We found a total of 78 ligand–receptor interaction pairs (Figure5A). Based on pathway analysis, these ligand–receptor interactions were enriched among Rap1 signaling path- way (FDR = 6.1 × 10−12), Ras signaling pathway (FDR = 1.4 × 10−10), PI3K-Akt signaling pathway (FDR = 1.0 × 10−8), Hippo signaling pathway (FDR = 1.6 × 10−4), MAPK signal- ing pathway (FDR = 0.0014), Wnt signaling pathway (FDR = 0.0031), cytokine–cytokine receptor interaction (FDR = 0.0039), regulation of actin cytoskeleton (FDR = 0.028) and endocytosis (FDR = 0.049) (Figure5B). Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 6 of 13

Int.Int. J. J.Mol. Mol. Sci. Sci. 20212021, 22, 22, x, 5177FOR PEER REVIEW 66 of of 13 13

Figure 4. A single-cell atlas of E4.5 embryos: (A) TSNE clustering of single cells from mouse E4.5 blastocysts which were collected from GD5 uteri from a previous study. EPI: epiblast; PE: primitive endoderm; mTE: mural trophectoderm; pTE: polar trophectoderm. (B–D) TSNE plots showing the expression pattern of canonical marker genes for EPI (B), PE (C) and mTE/pTE (D). Dashed lines show the boundaries of the specific cell clusters.

During mouse embryo implantation, the blastocysts attach to the LE of the uterus in the way of mTE. Thus, we used CellPhoneDB (https://www.cellphonedb.org/ (accessed 22 April 2021)) to predict the ligand–receptor interactions between LE and mTE. We found a total of 78 ligand–receptor interaction pairs (Figure 5A). Based on pathway analysis, these ligand–receptor interactions were enriched among Rap1 signaling pathway (FDR = 6.1 × 10−12), Ras signaling pathway (FDR = 1.4 × 10−10), PI3K-Akt signaling pathway FigureFigure 4. 4. AA single-cell single-cell atlas atlas(FDR of of E4.5 E4.5 = 1.0embryos: embryos: × 10− 8(), (AA Hippo)) TSNE TSNE clustering clusteringsignaling ofpathway single cellscells (FDR fromfrom = mouse mouse1.6 × E4.510E4.5−4), blastocystsblastocysts MAPK signaling which which were were pathway collectedcollected from from GD5 GD5 uteri uteri from from(FDR a a previous= previous 0.0014), study. study. Wnt EPI: EPI:signaling epibla epiblast;st; pathway PE: primitive (FDR endoderm; = 0.0031), mTE:mTE: cytokine–cytokine muralmural trophectoderm;trophectoderm; receptor pTE: pTE: interac- polarpolar trophectoderm. trophectoderm. (B (B––DDtion)) TSNE TSNE (FDR plots plots = showing0.0039), showing regulationthe the expression expression of patternactin cytoskeleton ofof canonicalcanonical markermarker (FDR genes =genes 0.028) for for EPIand EPI ( B(endocytosisB),), PE PE ( C(C)) and and (FDR = mTE/pTEmTE/pTE (D (D). ).Dashed Dashed lines lines0.049) show show (Figurethe the boundaries boundaries 5B). of of the the specific specific cell cell clusters. clusters. During mouse embryo implantation, the blastocysts attach to the LE of the uterus in the way of mTE. Thus, we used CellPhoneDB (https://www.cellphonedb.org/ (accessed 22 April 2021)) to predict the ligand–receptor interactions between LE and mTE. We found a total of 78 ligand–receptor interaction pairs (Figure 5A). Based on pathway analysis, these ligand–receptor interactions were enriched among Rap1 signaling pathway (FDR = 6.1 × 10−12), Ras signaling pathway (FDR = 1.4 × 10−10), PI3K-Akt signaling pathway (FDR = 1.0 × 10−8), Hippo signaling pathway (FDR = 1.6 × 10−4), MAPK signaling pathway (FDR = 0.0014), Wnt signaling pathway (FDR = 0.0031), cytokine–cytokine receptor interac- tion (FDR = 0.0039), regulation of actin cytoskeleton (FDR = 0.028) and endocytosis (FDR = 0.049) (Figure 5B).

FigureFigure 5. Cell–cell 5. Cell–cell communication communication between between mTE mTE and LE: and ( LE:A) Dot (A) plot Dot showing plot showing selected selected ligand–receptor ligand–receptor interactions interactions underly- ingunderlying mTE-LE crosstalk. mTE-LE p crosstalk.-values arep-values indicated are indicatedby circle size by circle and sizemeans and of means the average of the averageexpression expression level of levelinteracting of interacting molecule aremolecule indicated areby color; indicated (B) KEGG by color; Pathway (B) KEGG enrichment Pathway analysis enrichment of ligand–receptor analysis of ligand–receptor pairs by using the pairs DAVID by using online the tools. DAVID online tools. 2.5. Inferring Uterine Epithelial-Stromal Crosstalk at the Implantation Site 2.5.It Inferring has been Uterine well Epithelial-Stromalestablished that uterine Crosstalk epithelial-stromal at the Implantation Sitecrosstalk is important for embryoIt hasimplantation been well established[24]. In our thatsingle-cell uterine RNA-seq epithelial-stromal data, we crosstalk identified is importantfour clusters for of embryo implantation [24]. In our single-cell RNA-seq data, we identified four clusters of stromal cells: S1, S1p, S2 and S2p. Their relative abundance was 36.5%, 19.0%, 34.6%, 9.9% at IS, and 63.1%, 1.6%, 32.1%, 3.2% at IIS (Figure6A). By analyzing the gene signature for Figure 5. Cell–cell communicationeach between cell cluster, mTE weand confirmed LE: (A) Dot that plot S1pshowing was selected related ligand–receptor to S1 and S2p interactions was related underly- to S2. S1p ing mTE-LE crosstalk. p-valuesand are S2p indicated were proliferatingby circle size and subsets means of of S1 th ande average S2, respectively expression level (Figure of interacting6B). molecule are indicated by color; (B) KEGG PathwayTo further enrichment reveal theanalysis relationship of ligand–receptor between pairs these by four using stromal the DAVID cell clusters, online tools. pseudotime trajectory analysis was conducted. Cells were arranged in a pseudotime manner with a 2.5.pedigree Inferring reconstruction Uterine Epithelial-Stroma algorithm forl biologicalCrosstalk at processes the Implantation based on Site transcriptional similar- ity. WeIt has found been that well all established stromal cell that clusters uterine formed epithelial-stromal a continuous crosstalk trajectory is (Figure important7A–C for). embryo implantation [24]. In our single-cell RNA-seq data, we identified four clusters of Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 7 of 13

stromal cells: S1, S1p, S2 and S2p. Their relative abundance was 36.5%, 19.0%, 34.6%, 9.9% at IS, and 63.1%, 1.6%, 32.1%, 3.2% at IIS (Figure 6A). By analyzing the gene signa- ture for each cell cluster, we confirmed that S1p was related to S1 and S2p was related to S2. S1p and S2p were proliferating subsets of S1 and S2, respectively (Figure 6B).

Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 7 of 13

Int. J. Mol. Sci. 2021, 22, 5177 7 of 13

stromal cells: S1, S1p, S2 and S2p. Their relative abundance was 36.5%, 19.0%, 34.6%, 9.9%We mappedat IS, and three 63.1%, marker 1.6%, genes, 32.1%, namely 3.2% at Hand2, IIS (Figure Hoxa11 6A). and By Mki67,analyzing on thethe trajectorygene signa- ture(Figure for each7D). Ourcell cluster, results suggestedwe confirmed a S2p that→ S1pS2 → wasS1 related→ S1p trajectory:to S1 and S2p (1) S2pwas wasrelated the to S2.origin S1p and of all S2p stromal were cells; proliferating (2) S2 was subsets able to of transit S1 and to S1;S2, and respectively (3) S1p was (Figure derived 6B). from S1.

Figure 6. Sub-clusters of stromal cells: (A) TSNE plot showing four clusters of stromal cells. Relative cell type abundance was shown as insets; (B) heatmap of top 10 gene expression signatures for each cell type.

To further reveal the relationship between these four stromal cell clusters, pseudo- time trajectory analysis was conducted. Cells were arranged in a pseudotime manner with a pedigree reconstruction algorithm for biological processes based on transcrip- tional similarity. We found that all stromal cell clusters formed a continuous trajectory (Figure 7A–C). We mapped three marker genes, namely Hand2, Hoxa11 and Mki67, on the trajectory (Figure 7D). Our results suggested a S2p → S2 → S1 → S1p trajectory: FigureFigure 6. Sub-clusters 6. Sub-clusters of ofstromal stromal(1) S2p cells: cells: was (A ( Athe) )TSNE TSNE origin plot plot of showing showing all stromal four clustersclusterscells; (2) ofof stromalS2 stromal was cells.able cells. Relativeto Relative transit cell cell to type S1;type abundance and abundance (3) S1p was waswas shown shown as asinsets; insets; (B ()B heatmap) heatmapderived of of topfrom top 10 10 S1. gene gene expression expression signaturessignatures for for each each cell cell type. type.

To further reveal the relationship between these four stromal cell clusters, pseudo- time trajectory analysis was conducted. Cells were arranged in a pseudotime manner with a pedigree reconstruction algorithm for biological processes based on transcrip- tional similarity. We found that all stromal cell clusters formed a continuous trajectory (Figure 7A–C). We mapped three marker genes, namely Hand2, Hoxa11 and Mki67, on the trajectory (Figure 7D). Our results suggested a S2p → S2 → S1 → S1p trajectory: (1) S2p was the origin of all stromal cells; (2) S2 was able to transit to S1; and (3) S1p was derived from S1.

Figure 7. Pseudotime ordering of stromal cells: (A–C) The distribution of pseudotime (A), cell origin (B) and cell type (C) across the reconstructed trajectory. The numbers inside the black circles represent the different cell status numbers identified in the trajectory analysis; (D) line plots showing gene expression dynamics of Hand2, Hoxa11 and Mki67 in the pseudotime order. Upon embryo implantation, stromal cells proximally surrounding the implantation chamber (i.e., S1p cells) exhibit proliferation and spreading. These cells differentiate and form the primary decidual zone (PDZ) on the afternoon of gestational day 5 [25,26]. Thus,

Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 8 of 13

Figure 7. Pseudotime ordering of stromal cells: (A–C) The distribution of pseudotime (A), cell origin (B) and cell type (C) across the reconstructed trajectory. The numbers inside the black circles represent the different cell status numbers iden- tified in the trajectory analysis; (D) line plots showing gene expression dynamics of Hand2, Hoxa11 and Mki67 in the pseudotime order.

Int. J. Mol. Sci. 2021, 22, 5177 Upon embryo implantation, stromal cells proximally surrounding the implantation8 of 13 chamber (i.e., S1p cells) exhibit proliferation and spreading. These cells differentiate and form the primary decidual zone (PDZ) on the afternoon of gestational day 5 [25,26]. Thus, the interaction between LE and S1p represents the uterine epithelial-stromal crosstalk. By the interaction between LE and S1p represents the uterine epithelial-stromal crosstalk. usingBy using CellPhoneDB, CellPhoneDB, we predicted we predicted the theligand–receptor ligand–receptor interactions interactions between between LE LE and and S1p. WeS1p. identified We identified a total a of total 77 ofligand–receptor 77 ligand–receptor interaction interaction pairs pairs (Figure (Figure 8A).8A). Based Based on onpath- waypathway analysis, analysis, these these ligand–receptor ligand–receptor interactions interactions were were enriched amongamong the the PI3K-Akt PI3K-Akt signalingsignaling pathway pathway (FDR (FDR = = 2.9 2.9 ×× 1010−15−),15 ECM-receptor), ECM-receptor interaction interaction (FDR (FDR = =1.4 1.4 × ×10−109),− Rap19), signalingRap1 signaling pathway pathway (FDR (FDR= 1.4 =× 1.410−8×), 10Ras−8 ),signaling Ras signaling pathway pathway (FDR (FDR = 2.9 = × 2.9 10×−7),10 Hippo−7), signalingHippo signaling pathway pathway (FDR = (FDR2.8 × 10 = 2.8−5), ×Wnt10 −signaling5), Wnt signaling pathway pathway (FDR = (FDR7.3 × 10 = 7.3−4) and× 10 HIF−4) −1 signalingand HIF −pathway1 signaling (FDR pathway = 0.026) (FDR (Figure = 0.026) 8B). (Figure 8B).

FigureFigure 8. Cell–cell 8. Cell–cell communication communication between between LE LE and and S1p: S1p: (A) ( Adot) dot plot plot showing showing selected selected ligand–receptor ligand–receptor interactions interactions un- derlyingunderlying LE-S1p LE-S1p crosstalk; crosstalk; (B) KEGG (B) KEGG Pathway Pathway enrichment enrichment analysis analysis of ofligand–receptor ligand–receptor pairs. pairs.

3.3. Discussion Discussion TheThe mouse mouse is is widely widely used used as as an an animal animal model model for for investigating investigating human human embryo embryo im- plantation.implantation. In this In thisstudy, study, by profiling by profiling the the single-cell single-cell transcriptome transcriptome for for 8421 8421 cells cells using using the 10xthe Genomics 10x Genomics approach, approach, we revealed we revealed 19 distinct 19 distinct cell cell clusters, clusters, including including four four stromal stromal cell clusters,cell clusters, two epithelial two epithelial cell clusters, cell clusters, one smooth one smooth muscle muscle cell cluster, cell cluster, one pericyte one pericyte cluster, fourcluster, endothelial four endothelial cell clusters cell clusters and seven and sevenimmu immunene cell clusters cell clusters at the at the mouse mouse embryo embryo im- plantationimplantation site. site. To the To thebest best of our of our knowledge, knowledge, the the present present study study is isthe the first first to to highlight highlight the transcriptomethe transcriptome landscape landscape of embryo of embryo impl implantationantation at single-cell at single-cell resolution. resolution. In our single-cell RNA-seq data, the cell type composition for the implantation site In our single-cell RNA-seq data, the cell type composition for the implantation site of the uterus is 3.2% epithelial cells, 44.9% stromal cells, 1.8% smooth muscle cells, 3.9% of the uterus is 3.2% epithelial cells, 44.9% stromal cells, 1.8% smooth muscle cells, 3.9% pericytes, 12.8% endothelial cells and 33.4% immune cells, while the cell type composition pericytes,for the inter-implantation 12.8% endothelial site cells of the and uterus 33.4% is 4.2% immune epithelial cells, cells, while 41.4% the stromal cell type cells, composi- 1.8% tionsmooth for the muscle inter-implantation cells, 5.7% pericytes, site of 12.3% the endothelialuterus is 4.2% cells epithelial and 20.1% cells, immune 41.4% cells. stromal By cells,using 1.8% the criteriasmooth ofmusclep < 0.05 cells, and 5.7% fold changepericytes, >2, 12.3% the proportions endothelial of allcells cell and types 20.1% were im- muneunchanged cells. By except using for the LEC criteria and Bof cells. p < 0.05 In fact, and LEC fold cells change and >2, B cells the areproportions minor cell of types all cell types(<5%) were in the unchanged uterus, and except thus thefor estimatedLEC and B percentages cells. In fact, for LEC these cells two and cell typesB cells might are minor be inaccurate. These results were in line with the fact that there are only minimal histological changes at the implantation site compared to the inter-implantation site on gestational day 5 [6]. We investigated the breadth of transcriptional changes for each cell type by perform- ing differential gene expression analysis. S1p cells have the largest number of differentially Int. J. Mol. Sci. 2021, 22, 5177 9 of 13

expressed genes. In this study, we identified four stromal cell clusters: S1, S1p, S2 and S2p. S1 and its proliferating subset S1p were superficial stromal cells expressing Hand2, while S2 and its proliferating subset S2p were deep stromal cells which were negative for Hand2. Upon embryo implantation, stromal cells proximally surrounding the implantation chamber (i.e., S1p cells) exhibit proliferation and spreading. These cells differentiate and form the primary decidual zone (PDZ) on the afternoon of gestational day 5 [25,26]. Gene ontology analysis showed that stress response, cell death, transport, cell cycle and prolif- eration, cell organization and biogenesis, developmental processes, metabolism, DNA metabolism and RNA metabolism were significantly enriched among differentially expressed genes. We were also interested in genes differentially expressed in LE. Blastocyst attachment to the uterine LE occurs at midnight of gestational day 4 and a firm connection between blastocyst attachment to the uterine LE is established on the morning of gesta- tional day 5 [27]. We identified 391 differentially expressed genes in LE, of which 132 genes were upregulated and 259 genes were downregulated in IS compared to IIS. Gene ontology analysis revealed that cell adhesion, stress response, cell death, cell cycle and proliferation, cell organization and biogenesis, developmental processes, protein metabolism and RNA metabolism were significantly enriched among differentially expressed genes. Considering spatial relationships between cell types at the implantation site, LE and S1p cells are likely the top two contributors to embryo implantation in the uterus. Nevertheless, we would like to note that immune cells (especially M and DC) [28], as well as GE [29], may also largely contribute to embryo implantation according to the literature. Previously, the E4.5 blastocysts obtained from GD5 uteri have been subjected to single-cell RNA-seq [23]. Through data integration, we inferred cell–cell communication between E4.5 blastocysts and the implantation site of GD5 uteri by their expression of ligand–receptor pairs. We were particularly interested in the interactions between mTE from the blastocyst and LE from the uterus, as physical contact between these two cell types represents the key mechanism of embryo implantation. Notably, LE cells expressed FGF9, while the corresponding receptors (FGFR1, FGFR3 and FGFR4) were expressed in mTE. On the other hand, mTE cells expressed FGF4 and FGF10, while the correspond- ing receptor FGFR2 was expressed in mTE. These data highlight the importance of FGF signaling in embryo implantation. The FGF signaling is part of the Rap1 signaling path- way, Ras signaling pathway, PI3K-Akt signaling pathway, MAPK signaling pathway and cytokine-cytokine receptor interaction. Additionally, we found that Wnt signaling pathway and Hippo signaling pathway might also play a significant role in LE-mTE interaction. Previous studies have shown that uterine epithelial-stromal crosstalk is crucial for embryo implantation as well as post-implantation development of the uterus [24]. In this study, we narrowed down uterine epithelial-stromal crosstalk to the interaction between LE and S1p cells. We found that FGF and Wnt signaling also play an important role in LE-S1p interaction. Although the interactions between LE/S1p and immune cells might also contribute to embryo implantation, they were not pursued in this study. In conclusion, this study provided a comprehensive single-cell transcriptome atlas for the mouse embryo implantation site. Our data present a valuable resource for deciphering the molecular mechanism underlying embryo implantation.

4. Materials and Methods 4.1. Sample Collection Adult SPF-grade CD-1 mice were used in this study. All mice were caged under light-controlled conditions with 14 h/10 h light/dark cycles and free access to regular food and water. Female mice were mated with fertile males and the mating was confirmed the next morning by the presence of a vaginal plug. The day of the vaginal plug was denoted as gestation day 1 (GD1). On the morning of GD5, embryo implantation positions along the uterus were identified after an intravenous injection of Chicago Blue B dye solution. The whole uterus segments for implantation sites (IS) and inter-implantation sites (IIS) were Int. J. Mol. Sci. 2021, 22, 5177 10 of 13

collected separately. All animal procedures were approved by the Institutional Animal Care and Use Committee of South China Agricultural University (Guangzhou, China).

4.2. Single-Cell Dissociation of Mouse Uterus Whole uterus segments were incubated in the dissociation buffer containing 2 mg/mL Collagenase II (#C6885, Sigma-Aldrich, St. Louis, MO, USA), 10 mg/mL Dispase II (#354235, Corning, Corning, NY, USA) and 50,000 U/mL DNase I (#DN25, Sigma-Aldrich, St. Louis, MO, USA) for up to 30 min at 37 ◦C in a shaking incubator. The digestion progress was checked every 5 min with a microscope until single-cell suspension was achieved. The single-cell suspension was then passed through a 40-µm cell strainer to remove undigested tissues. Cells were spun down at 250× g at 4 ◦C for 4 min and the pelleted cells were washed using centrifugation. In order to measure cell viability, cells were strained with AO/PI solution (#CS2-0106, Nexcelom Bioscience, Lawrence, MA, USA) and counted using a Cellometer Auto 2000 instrument (#SD-100, Nexcelom Bioscience, Lawrence, MA, USA). The single-cell suspension was carried forward to single-cell RNA-seq only if the cell viability was >80% and the percentage of cell clumps was <10%.

4.3. Single-Cell RNA-Seq Library Preparation and Sequencing The final concentration of single-cell suspension was adjusted to 1000 cells/µL and a volume of 15 µL was loaded into one channel of the ChromiumTM Single Cell B Chip (#1000073, 10x Genomics, Pleasanton, CA, USA), aiming at recovering 8000–10,000 cells. The Chromium Single Cell 30 Library and Gel Bead Kit v3 (#1000075, 10x Genomics) was used for single-cell bar-coding, cDNA synthesis and library preparation, following the manufacturer0s instructions provided as the Single Cell 30 Reagent Kits User Guide Version 3. Library sequencing was performed on a NovaSeq 6000 system (Illumina, San Diego, CA, USA) configured with the paired-end 150-bp protocol at a sequencing depth of approximately 400 million reads.

4.4. Single-Cell RNA-Seq Data Analysis Raw data of bcl files from the NovaSeq 6000 system were converted to fastq files using the bcl2fastq2 tool v2.19.0.316 (Illumina, San Diego, CA, USA). These fastq files were aligned to the 10 mm mouse reference genome by using the CellRanger software v3.0.1 (10x Genomics, Pleasanton, CA, USA). The resulting gene count matrix was processed with the R package Seurat v3.1.3 [30]. Cells with fewer than 200 or greater than 6000 unique genes, as well as cells with greater than 25% mitochondrial counts, were excluded. Meanwhile, genes expressed in fewer than 3 cells were removed. Following data filtering, the clean gene count matrix was normalized and scaled by using NormalizeData and ScaleData, respectively. The top 2000 highest variable genes were used for the principal component analysis (PCA) and the optimal number of PCA components was determined by the JackStraw procedure. Single cells were clustered by the graph-based algorithm in PCA space and visualized using the t-distributed stochastic neighbor embedding (tSNE) dimensional reduction technique. The cell type label for each cell cluster was manually assigned based on canonical cell markers. The FindAllMarkers function was used to identify novel marker genes for each cluster with a minimum of 20% of cells expressing the gene within the cluster and a minimum logFC threshold of 0.25. In order to find differentially expressed genes in the same cell type between pre-receptive uteri and receptive uteri, the FindMarkers function in Seurat was used with min.pct being set to 0.20 and min.logfc being set to 0.25.

4.5. Gene Ontology Analysis Gene ontology (GO) analysis was performed as described previously [31]. GO terms were grouped according to the biological process category in the Mouse Genome In- formatics (MGI) GOslim database [32]. To test for enrichment, a hypergeometric test was conducted and p < 0.05 was used as the significance threshold to identify enriched GO terms. Int. J. Mol. Sci. 2021, 22, 5177 11 of 13

4.6. Pathway Enrichment Analysis Pathway enrichment analysis was conducted by using the DAVID online tools 6.8 [33]. The significance threshold for FDR was set at 0.05.

4.7. Cell–Cell Communication Analysis The single-cell RNA-seq data for mouse E4.5 blastocysts were downloaded from the GEO database (GSE63266) [23] and re-analyzed with the same pipeline as described above. E4.5 is equivalent to GD5. In order to analyze cell–cell communication between blastocysts and uteri, we merged E4.5 blastocyst data and uterus IS data into a single Seurat object, from which a meta file as well as a count file was generated. These 2 files were used as input for the CellphoneDB software v2.1.4 [34] to infer cell–cell communication based on ligand–receptor interactions with default parameters. p < 0.05 were considered statistically significant.

4.8. Pseudotime Analysis The Monocle2 package v2.18.0 was used for pseudotime analysis [35]. The count data and meta data were exported from the Seurat object and then imported into the CellDataSet object in Monocle2. Feature genes were selected by using the differentialGeneTest function. After reducing dimensions using the DDRTree algorithm, the orderCells function was used to infer the trajectory with default parameters. The reconstructed trajectory was visualized by the plot_cell_trajectory function. The plot_genes_in_pseudotime function was used to map gene expression dynamics.

Supplementary Materials: The following are available online at https://www.mdpi.com/article/10 .3390/ijms22105177/s1. Author Contributions: J.-L.L. supervised the study. J.-L.L. designed the experiments. Y.Y. and J.-P.H. performed the experiments. Y.Y. and J.-L.L. analyzed the data. Y.Y. and J.-L.L. wrote the paper. All authors have read and agreed to the published version of the manuscript. Funding: This research was funded by the National Natural Science Foundation of China (32070845 and 31771665), the Guangdong Natural Science Funds for Distinguished Young Scholars (2021B1515020079), and the Innovation Team Project of Guangdong University (2019KCXTD001). Institutional Review Board Statement: All animal procedures were approved by the Institutional Animal Care and Use Committee of South China Agricultural University (No. 2020B078 approved on 29/09/2020). Informed Consent Statement: Not applicable. Data Availability Statement: The data presented in this study are available on request from the corresponding author. Conflicts of Interest: The authors declare no conflict of interest.

References 1. Wilcox, L.S.; Peterson, H.B.; Haseltine, F.P.; Martin, M.C. Defining and interpreting pregnancy success rates for in vitro fertilization. Fertil. Steril. 1993, 60, 18–25. [CrossRef] 2. Zinaman, M.J.; Clegg, E.D.; Brown, C.C.; O’Connor, J.; Selevan, S.G. Estimates of human fertility and pregnancy loss. Fertil. Steril. 1996, 65, 503–509. [CrossRef] 3. Wang, H.; Dey, S.K. Roadmap to embryo implantation: Clues from mouse models. Nat. Rev. Genet. 2006, 7, 185–199. [CrossRef] 4. Zhang, S.; Lin, H.; Kong, S.; Wang, S.; Wang, H.; Wang, H.; Armant, D.R. Physiological and molecular determinants of embryo implantation. Mol. Asp. Med. 2013, 34, 939–980. [CrossRef][PubMed] 5. Cha, J.; Sun, X.; Dey, S.K. Mechanisms of implantation: Strategies for successful pregnancy. Nat. Med. 2012, 18, 1754–1767. [CrossRef] 6. Ma, X.H.; Hu, S.J.; Ni, H.; Zhao, Y.C.; Tian, Z.; Liu, J.L.; Ren, G.; Liang, X.H.; Yu, H.; Wan, P.; et al. Serial analysis of gene expression in mouse uterus at the implantation site. J. Biol. Chem. 2006, 281, 9351–9360. [CrossRef] Int. J. Mol. Sci. 2021, 22, 5177 12 of 13

7. Reese, J.; Das, S.K.; Paria, B.C.; Lim, H.; Song, H.; Matsumoto, H.; Knudtson, K.L.; DuBois, R.N.; Dey, S.K. Global gene expression analysis to identify molecular markers of uterine receptivity and embryo implantation. J. Biol. Chem. 2001, 276, 44137–44145. [CrossRef] 8. Zhang, W.Q.; Zhao, M.; Huang, M.Y.; Liu, J.L. Comparative transcriptomic analysis of embryo implantation in mice and rats. Cell. Physiol. Biochem. 2018, 50, 668–678. [CrossRef][PubMed] 9. Chen, Y.; Ni, H.; Ma, X.H.; Hu, S.J.; Luan, L.M.; Ren, G.; Zhao, Y.C.; Li, S.J.; Diao, H.L.; Xu, X.; et al. Global analysis of differential luminal epithelial gene expression at mouse implantation sites. J. Mol. Endocrinol. 2006, 37, 147–161. [CrossRef][PubMed] 10. Yoon, S.J.; Choi, D.H.; Lee, W.S.; Cha, K.Y.; Kim, S.N.; Lee, K.A. A molecular basis for embryo apposition at the luminal epithelium. Mol. Cell. Endocrinol. 2004, 219, 95–104. [CrossRef] 11. Svensson, V.; Vento-Tormo, R.; Teichmann, S.A. Exponential scaling of single-cell rna-seq in the past decade. Nat. Protoc. 2018, 13, 599–604. [CrossRef][PubMed] 12. Gendron, R.L.; Paradis, H.; Hsieh-Li, H.M.; Lee, D.W.; Potter, S.S.; Markoff, E. Abnormal uterine stromal and glandular function associated with maternal reproductive defects in hoxa-11 null mice. Biol. Reprod. 1997, 56, 1097–1105. [CrossRef] 13. Reardon, S.N.; King, M.L.; MacLean, J.A., 2nd; Mann, J.L.; DeMayo, F.J.; Lydon, J.P.; Hayashi, K. Cdh1 is essential for endometrial differentiation, gland development, and adult function in the mouse uterus. Biol. Reprod. 2012, 86, 141. [CrossRef][PubMed] 14. Suryawanshi, H.; Morozov, P.; Straus, A.; Sahasrabudhe, N.; Max, K.E.A.; Garzia, A.; Kustagi, M.; Tuschl, T.; Williams, Z. A single-cell survey of the human first-trimester placenta and decidua. Sci. Adv. 2018, 4, eaau4788. [CrossRef][PubMed] 15. Mucenski, M.L.; Mahoney, R.; Adam, M.; Potter, A.S.; Potter, S.S. Single cell rna-seq study of wild type and hox9,10,11 mutant developing uterus. Sci. Rep. 2019, 9, 4557. [CrossRef][PubMed] 16. Kalucka, J.; de Rooij, L.; Goveia, J.; Rohlenova, K.; Dumas, S.J.; Meta, E.; Conchinha, N.V.; Taverna, F.; Teuwen, L.A.; Veys, K.; et al. Single-cell transcriptome atlas of murine endothelial cells. Cell 2020, 180, 764–779 e720. [CrossRef] 17. Croy, B.A.; Chen, Z.; Hofmann, A.P.; Lord, E.M.; Sedlacek, A.L.; Gerber, S.A. Imaging of vascular development in early mouse decidua and its association with leukocytes and trophoblasts. Biol. Reprod. 2012, 87, 125. [CrossRef] 18. Li, Q.; Kannan, A.; DeMayo, F.J.; Lydon, J.P.; Cooke, P.S.; Yamagishi, H.; Srivastava, D.; Bagchi, M.K.; Bagchi, I.C. The antiproliferative action of progesterone in uterine epithelium is mediated by hand2. Science 2011, 331, 912–916. [CrossRef] 19. Fu, D.J.; De Micheli, A.J.; Bidarimath, M.; Ellenson, L.H.; Cosgrove, B.D.; Flesken-Nikitin, A.; Nikitin, A.Y. Cells expressing pax8 are the main source of homeostatic regeneration of adult mouse endometrial epithelium and give rise to serous endometrial carcinoma. Dis. Model. Mech. 2020, 13.[CrossRef] 20. Keenihan, S.N.; Robertson, S.A. Diversity in phenotype and steroid hormone dependence in dendritic cells and macrophages in the mouse uterus. Biol. Reprod. 2004, 70, 1562–1572. [CrossRef] 21. Ye, Y.; Gaugler, B.; Mohty, M.; Malard, F. Plasmacytoid dendritic cell biology and its role in immune-mediated diseases. Clin. Transl. Immunol. 2020, 9, e1139. [CrossRef][PubMed] 22. Hilton, H.G.; Rubinstein, N.D.; Janki, P.; Ireland, A.T.; Bernstein, N.; Fong, N.L.; Wright, K.M.; Smith, M.; Finkle, D.; Martin- McNulty, B.; et al. Single-cell transcriptomics of the naked mole-rat reveals unexpected features of mammalian immunity. PLoS Biol. 2019, 17, e3000528. [CrossRef][PubMed] 23. Nakamura, T.; Yabuta, Y.; Okamoto, I.; Aramaki, S.; Yokobayashi, S.; Kurimoto, K.; Sekiguchi, K.; Nakagawa, M.; Yamamoto, T.; Saitou, M. Sc3-seq: A method for highly parallel and quantitative measurement of single-cell gene expression. Nucleic Acids Res. 2015, 43, e60. [CrossRef][PubMed] 24. Hantak, A.M.; Bagchi, I.C.; Bagchi, M.K. Role of uterine stromal-epithelial crosstalk in embryo implantation. Int. J. Dev. Biol. 2014, 58, 139–146. [CrossRef] 25. Li, Y.; Dewar, A.; Kim, Y.S.; Dey, S.K.; Sun, X. Pregnancy success in mice requires appropriate cannabinoid receptor signaling for primary decidua formation. Elife 2020, 9, e61762. [CrossRef][PubMed] 26. Yuan, J.; Aikawa, S.; Deng, W.; Bartos, A.; Walz, G.; Grahammer, F.; Huber, T.B.; Sun, X.; Dey, S.K. Primary decidual zone formation requires scribble for pregnancy success in mice. Nat. Commun. 2019, 10, 5425. [CrossRef] 27. Ye, X. Uterine luminal epithelium as the transient gateway for embryo implantation. Trends Endocrinol. Metab. 2020, 31, 165–180. [CrossRef] 28. Zenclussen, A.C.; Hammerling, G.J. Cellular regulation of the uterine microenvironment that enables embryo implantation. Front. Immunol. 2015, 6, 321. [CrossRef] 29. Kelleher, A.M.; DeMayo, F.J.; Spencer, T.E. Uterine glands: Developmental biology and functional roles in pregnancy. Endocr. Rev. 2019, 40, 1424–1445. [CrossRef][PubMed] 30. Stuart, T.; Butler, A.; Hoffman, P.; Hafemeister, C.; Papalexi, E.; Mauck, W.M., 3rd; Hao, Y.; Stoeckius, M.; Smibert, P.; Satija, R. Comprehensive integration of single-cell data. Cell 2019, 177, 1888–1902.e1821. [CrossRef] 31. Liu, J.L.; Zuo, R.J.; Peng, Y.; Fu, Y.S. The impact of multiparity on uterine gene expression and decidualization in mice. Reprod. Sci. 2016, 23, 687–694. [CrossRef][PubMed] 32. Law, M.; Shaw, D.R. Mouse genome informatics (MGI) is the international resource for information on the . Methods Mol. Biol. 2018, 1757, 141–161. [PubMed] 33. Huang, D.W.; Sherman, B.T.; Tan, Q.; Kir, J.; Liu, D.; Bryant, D.; Guo, Y.; Stephens, R.; Baseler, M.W.; Lane, H.C.; et al. David bioinformatics resources: Expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 2007, 35, W169–W175. [CrossRef] Int. J. Mol. Sci. 2021, 22, 5177 13 of 13

34. Efremova, M.; Vento-Tormo, M.; Teichmann, S.A.; Vento-Tormo, R. Cellphonedb: Inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat. Protoc. 2020, 15, 1484–1506. [CrossRef] 35. Qiu, X.; Hill, A.; Packer, J.; Lin, D.; Ma, Y.A.; Trapnell, C. Single-cell mrna quantification and differential analysis with census. Nat. Methods 2017, 14, 309–315. [CrossRef][PubMed]