Recurrent pregnancy loss is associated with a pro-senescent decidual response during the peri-implantation window

Emma S Lucas, Pavle Vrljicak, Joanne Muter, Maria M Diniz-da-Costa, Paul J Brighton, Chow-Seng Kong, Julia Lipecki, Katherine J Fishwick, Joshua Odendaal, Lauren J Ewington, Siobhan Quenby, Sascha Ott, Jan J Brosens, Emma S Lucas, Pavle Vrljicak, Joanne Muter, Maria M Diniz-da-Costa, Paul J Brighton, Chow-Seng Kong, Julia Lipecki, Katherine J Fishwick, Joshua Odendaal, Lauren J Ewington, Siobhan Quenby, Sascha Ott, Jan J Brosens

Abstract

During the implantation window, the endometrium becomes poised to transition to a pregnant state, a process driven by differentiation of stromal cells into decidual cells (DC). Perturbations in this process, termed decidualization, leads to breakdown of the feto-maternal interface and miscarriage, but the underlying mechanisms are poorly understood. Here, we reconstructed the decidual pathway at single-cell level in vitro and demonstrate that stromal cells first mount an acute stress response before emerging as DC or senescent DC (snDC). In the absence of immune cell-mediated clearance of snDC, secondary senescence transforms DC into progesterone-resistant cells that abundantly express extracellular matrix remodelling factors. Additional single-cell analysis of midluteal endometrium identified DIO2 and SCARA5 as marker genes of a diverging decidual response in vivo. Finally, we report a conspicuous link between a pro-senescent decidual response in peri-implantation endometrium and recurrent pregnancy loss, suggesting that pre-pregnancy screening and intervention may reduce the burden of miscarriage.

Conflict of interest statement

The authors declare no competing non-financial interests but the following competing financial interests: Patent application, The University of Warwick, Professor Jan Brosens, application no. 1911947.8, pending, identification of SCARA5 and DIO2 as marker genes of decidual cells and senescent decidual cells, respectively. The remaining authors declare no competing financial interests.

Figures

Fig. 1. Single-cell reconstruction of the decidual…
Fig. 1. Single-cell reconstruction of the decidual pathway in culture.
a Schematic representation of in vitro time-course experiment. bt-SNE projection of 4580 EnSC, colour-coded according to days of decidualization (D0-8) and upon withdrawal (WD) of the differentiation signal for 48 h. D0 represents undifferentiated EnSC. c The same t-SNE plot now colour-coded according to transcriptional state (S1–7). d Violin plots showing log-transformed, normalized expression levels for indicated genes in decidual cells (DC, state S5) and senescent decidual cells (snDC, state S6). e Relative proportion of total cells assigned to states S5 (DC), S6 (snDC) and S7 at each experimental timepoint. f Decidualizing EnSC were placed in pseudotime to reconstruct the trajectory of differentiation, revealing a continuous trajectory towards senescence with a single branch point marking the divergence of DC. g Heatmap showing gene dynamics during cell state transition at the branch point shown in panel (f). Columns are points in pseudotime while rows represent the 50 most dynamic genes at the branch point. The beginning of pseudotime is in the middle of the heatmap and the trajectory towards DC and snDC are indicated by the arrows. Hierarchical clustering visualizes modules of genes with similar lineage-dependent expression patterns.
Fig. 2. K-means cluster analysis identifies co-regulated…
Fig. 2. K-means cluster analysis identifies co-regulated decidual gene networks.
Analysis of 1748 DEG across the decidual pathway (D0–D8) yielded seven networks of uniquely co-regulated genes. Networks are annotated for selected transcription factor genes with core roles in decidualization.
Fig. 3. Coordinated expression of decidual immune…
Fig. 3. Coordinated expression of decidual immune surveillance genes.
a Decidual gene network B2 annotated to highlight genes implicated in uNK-cell activation and immune surveillance of senescence cells. b Primary EnSC cultures were decidualized with 8-bromo-cAMP and MPA (C+M) for the indicated days. ELISAs were performed on spent medium collected at 48 h intervals to examine secreted levels of CXCL14, IL-15 and TIMP-3. Grey dotted lines indicate secreted levels in individual cultures (n = 4). Black solid lines indicate the median level of secretion. c Four independent primary EnSC cultures were decidualized with 8-bromo-cAMP and MPA (C+M) for the indicated days. ELISAs were performed on spent medium collected at 48 h intervals to examine secreted levels of clusterin (CLU) and sST2. Cultures established from the same biopsy are colour matched between plots (dotted lines). Black solid lines indicate the median level of secretion. d Schematic representation of uNK-cell co-culture experiments. A total of 5000 primary uNK cells were co-cultured for 48 h from the indicated timepoint with 50,000 decidualized cells seeded in 96-well plates. Four independent EnSC cultures were used. e SAβG activity in undifferentiated and decidualized (C+M) cells co-cultured with or without uNK cells. Four independent EnSC cultures were used. f Secretion of CLU in decidualized (C+M) cells co-cultured with uNK cells relative to decidualized cells cultured without uNK cells. g Secreted sST2 levels in the same culture. Four independent EnSC cultures were used; lines connect paired samples.
Fig. 4. Identification of endometrial cell types…
Fig. 4. Identification of endometrial cell types and subsets during the implantation window.
at-SNE plot of 2847 cells isolated from six LH-timed biopsies, coloured to indicate reported cycle day (LH + 8 or LH + 10), captures all major endometrial cell types, including epithelial cells (EpC), immune cells (IC), endothelial cells (EC), stromal cells (EnSC) and a discrete but transcriptionally distinct proliferative (PC) stromal subpopulation. EpC segregated in four subpopulations (EpC1-4). * indicates EpC3 contributed predominantly by a single sample. b additional dimensionality reduction separated immune cells into three uNK-cell subsets (NK1-3), naive B-cells (IC1), monocytes (IC2) and macrophage/dendritic cells (IC3). c Heatmap showing relative expression (z-score) of markers defining cell types and EpC subpopulations. MYO6, RASD1 and ALCAM are included as pan-epithelial genes. d Heatmap showing relative expression of markers defining endometrial IC populations during the implantation window, including three uNK-cell subsets.
Fig. 5. SCARA5 and DIO2 as marker…
Fig. 5. SCARA5 and DIO2 as marker genes for divergent decidual states.
aSCARA5 and DIO2 belong to two distinct decidual gene networks with peak expression in DC and snDC, respectively. b, c Spatial and temporal expression of SCARA5 and DIO2 in cycling human endometrium: b violin plots showing expression of SCARA5 and DIO2 in vivo in EnSC, EC (endothelial cells), EpC (epithelial cells) and IC (immune cells) (Wilcoxon rank sum test with Bonferroni correction); c expression of SCARA5 and DIO2 in proliferative and early-, mid-, late-luteal phase endometrium. Each bar represents an individual biopsy. The data were retrieved from microarray data deposited in the Gene Expression Omnibus (GEO Profiles, GDS2052). dSCARA5 and DIO2 transcript levels quantified by RT-qPCR analysis in 250 endometrial biopsies obtained between LH + 6 and LH + 11. Centile calculations were performed on dCt values using R software and graphs were generated based on the distribution of expression on each day. The median number of samples for each day was 43 (range: 30–46). e Multiplexed single-molecule in situ hybridization (smISH) on three endometrial biopsies (LH + 10) deemed SCARA5HIGH/DIO2LOW (95th and 20th percentile, respectively), SCARA5AVERAGE/DIO2AVERAGE (60th and 52th percentile, respectively), and SCARA5LOW/DIO2HIGH (4th and 91st percentile, respectively). Insert in the left panel shows hybridization with a negative control probe. Insert in middle panel shows SCARA5+ cells (blue, closed arrows) and DIO2+ cells (pink, open arrows) in close proximity. Scale bar: 100 µM. Original magnification: ×40.
Fig. 6. Impaired fate divergence of decidual…
Fig. 6. Impaired fate divergence of decidual cells in RPL.
a Distribution of uNK cells and SCARA5 and DIO2 percentiles in timed endometrial biopsies of control subjects (n = 90) and RPL patients (n = 89) (Welch two-sided t-test). b Number of RPL and control subjects across four bins defined by the sum of SCARA5 and uNK-cell percentiles. c Diagram illustrating the decidual pathway. Fate divergence of EnSC upon decidualization relates to the level of replicative stress (indicating by nuclear shading) incurred by individual cells during the proliferative phase. Stress-resistant decidual cells recruit and activate uNK cells to eliminate stressed/senescent decidual cells through granule exocytosis. Different defects along the decidual pathway can be identified, including ‘decidualization failure’, ‘excessive decidual senescence’ and ‘uNK cell deficiency’. The frequency of each defect in RPL and control subjects is shown (χ2 test).

References

    1. Rai R, Regan L. Recurrent miscarriage. Lancet. 2006;368:601–611. doi: 10.1016/S0140-6736(06)69204-0.
    1. Hardy K, Hardy PJ, Jacobs PA, Lewallen K, Hassold TJ. Temporal changes in chromosome abnormalities in human spontaneous abortions: results of 40 years of analysis. Am. J. Med Genet A. 2016;170:2671–2680. doi: 10.1002/ajmg.a.37795.
    1. Carp H, et al. Karyotype of the abortus in recurrent miscarriage. Fertil. Steril. 2001;75:678–682. doi: 10.1016/S0015-0282(00)01801-X.
    1. Stephenson MD, Awartani KA, Robinson WP. Cytogenetic analysis of miscarriages from couples with recurrent miscarriage: a case-control study. Hum. Reprod. 2002;17:446–451. doi: 10.1093/humrep/17.2.446.
    1. ESHRE. Recurrent Pregnancy Loss: A Guideline of the European Society of Human Reproduction and Embryology (2017).
    1. Practice Committee of the American Society for Reproductive, M. Evaluation and treatment of recurrent pregnancy loss. Fertil. Steril. 2012;5:1103–1111.
    1. Ogasawara M, Aoki K, Okada S, Suzumori K. Embryonic karyotype of abortuses in relation to the number of previous miscarriages. Fertil. Steril. 2000;73:300–304. doi: 10.1016/S0015-0282(99)00495-1.
    1. Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr. Rev. 2014;35:851–905. doi: 10.1210/er.2014-1045.
    1. Brighton, P. J. et al. Clearance of senescent decidual cells by uterine natural killer cells in cycling human endometrium. eLife6, e31274 (2017).
    1. Weimar CH, et al. Endometrial stromal cells of women with recurrent miscarriage fail to discriminate between high- and low-quality human embryos. PLoS One. 2012;7:e41424. doi: 10.1371/journal.pone.0041424.
    1. Brosens JJ, et al. Uterine selection of human embryos at implantation. Sci. Rep. 2014;4:3894. doi: 10.1038/srep03894.
    1. Brosens JJ, Pijnenborg R, Brosens IA. The myometrial junctional zone spiral arteries in normal and abnormal pregnancies: a review of the literature. Am. J. Obstet. Gynecol. 2002;187:1416–1423. doi: 10.1067/mob.2002.127305.
    1. Vrljicak P, et al. Analysis of chromatin accessibility in decidualizing human endometrial stromal cells. FASEB J. 2018;32:2467–2477. doi: 10.1096/fj.201701098R.
    1. Cloke B, et al. The androgen and progesterone receptors regulate distinct gene networks and cellular functions in decidualizing endometrium. Endocrinology. 2008;149:4462–4474. doi: 10.1210/en.2008-0356.
    1. Leitao B, et al. Silencing of the JNK pathway maintains progesterone receptor activity in decidualizing human endometrial stromal cells exposed to oxidative stress signals. FASEB J. 2010;24:1541–1551. doi: 10.1096/fj.09-149153.
    1. Muter J, et al. Progesterone-dependent induction of phospholipase C-related catalytically inactive protein 1 (PRIP-1) in decidualizing human endometrial stromal cells. Endocrinology. 2016;157:2883–2893. doi: 10.1210/en.2015-1914.
    1. Lynch VJ, Leclerc RD, May G, Wagner GP. Transposon-mediated rewiring of gene regulatory networks contributed to the evolution of pregnancy in mammals. Nat. Genet. 2011;43:1154–1159. doi: 10.1038/ng.917.
    1. Lynch VJ, et al. Ancient transposable elements transformed the uterine regulatory landscape and transcriptome during the evolution of mammalian pregnancy. Cell Rep. 2015;10:551–561. doi: 10.1016/j.celrep.2014.12.052.
    1. Erkenbrack EM, et al. The mammalian decidual cell evolved from a cellular stress response. PLoS Biol. 2018;16:e2005594. doi: 10.1371/journal.pbio.2005594.
    1. Al-Sabbagh M, et al. NADPH oxidase-derived reactive oxygen species mediate decidualization of human endometrial stromal cells in response to cyclic AMP signaling. Endocrinology. 2011;152:730–740. doi: 10.1210/en.2010-0899.
    1. Salker MS, et al. Disordered IL-33/ST2 activation in decidualizing stromal cells prolongs uterine receptivity in women with recurrent pregnancy loss. PLoS One. 2012;7:e52252. doi: 10.1371/journal.pone.0052252.
    1. Kajihara T, et al. Differential expression of FOXO1 and FOXO3a confers resistance to oxidative cell death upon endometrial decidualization. Mol. Endocrinol. 2006;20:2444–2455. doi: 10.1210/me.2006-0118.
    1. Kuroda K, et al. Induction of 11beta-HSD 1 and activation of distinct mineralocorticoid receptor- and glucocorticoid receptor-dependent gene networks in decidualizing human endometrial stromal cells. Mol. Endocrinol. 2013;27:192–202. doi: 10.1210/me.2012-1247.
    1. Antonangeli F, Zingoni A, Soriani A, Santoni A. Senescent cells: living or dying is a matter of NK cells. J. Leukoc. Biol. 2019;105:1275–1283. doi: 10.1002/JLB.MR0718-299R.
    1. Childs Bennett G., Gluscevic Martina, Baker Darren J., Laberge Remi-Martin, Marquess Dan, Dananberg Jamie, van Deursen Jan M. Senescent cells: an emerging target for diseases of ageing. Nature Reviews Drug Discovery. 2017;16(10):718–735. doi: 10.1038/nrd.2017.116.
    1. Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol. 2018;28:436–453. doi: 10.1016/j.tcb.2018.02.001.
    1. van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509:439–446. doi: 10.1038/nature13193.
    1. Jun JI, Lau LF. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 2010;12:676–685. doi: 10.1038/ncb2070.
    1. Storer M, et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell. 2013;155:1119–1130. doi: 10.1016/j.cell.2013.10.041.
    1. Ritschka B, et al. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes Dev. 2017;31:172–183. doi: 10.1101/gad.290635.116.
    1. Acosta JC, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133:1006–1018. doi: 10.1016/j.cell.2008.03.038.
    1. Ito Y, Hoare M, Narita M. Spatial and temporal control of senescence. Trends Cell Biol. 2017;27:820–832. doi: 10.1016/j.tcb.2017.07.004.
    1. Lucas ES, et al. Loss of endometrial plasticity in recurrent pregnancy loss. Stem Cells. 2016;34:346–356. doi: 10.1002/stem.2222.
    1. Macosko EZ, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015;161:1202–1214. doi: 10.1016/j.cell.2015.05.002.
    1. Kuroda K, et al. Elevated periimplantation uterine natural killer cell density in human endometrium is associated with impaired corticosteroid signaling in decidualizing stromal cells. J. Clin. Endocrinol. Metab. 2013;98:4429–4437. doi: 10.1210/jc.2013-1977.
    1. Song JJ, et al. Role of glutaredoxin in metabolic oxidative stress. Glutaredoxin as a sensor of oxidative stress mediated by H2O2. J. Biol. Chem. 2002;277:46566–46575. doi: 10.1074/jbc.M206826200.
    1. Zuo RJ, et al. Crystallin alphaB acts as a molecular guard in mouse decidualization: regulation and function during early pregnancy. FEBS Lett. 2014;588:2944–2951. doi: 10.1016/j.febslet.2014.05.045.
    1. Latini FR, et al. ABI3 ectopic expression reduces in vitro and in vivo cell growth properties while inducing senescence. BMC Cancer. 2011;11:11. doi: 10.1186/1471-2407-11-11.
    1. Michishita E, Garces G, Barrett JC, Horikawa I. Upregulation of the KIAA1199 gene is associated with cellular mortality. Cancer Lett. 2006;239:71–77. doi: 10.1016/j.canlet.2005.07.028.
    1. Petropoulou C, Trougakos IP, Kolettas E, Toussaint O, Gonos ES. Clusterin/apolipoprotein J is a novel biomarker of cellular senescence that does not affect the proliferative capacity of human diploid fibroblasts. FEBS Lett. 2001;509:287–297. doi: 10.1016/S0014-5793(01)03150-7.
    1. Trougakos IP. The molecular chaperone apolipoprotein J/clusterin as a sensor of oxidative stress: implications in therapeutic approaches—a mini-review. Gerontology. 2013;59:514–523. doi: 10.1159/000351207.
    1. Bianco AC, Kim BW. Deiodinases: implications of the local control of thyroid hormone action. J. Clin. Invest. 2006;116:2571–2579. doi: 10.1172/JCI29812.
    1. Severino V, et al. Insulin-like growth factor binding proteins 4 and 7 released by senescent cells promote premature senescence in mesenchymal stem cells. Cell Death Dis. 2013;4:e911. doi: 10.1038/cddis.2013.445.
    1. Le Maitre CL, Freemont AJ, Hoyland JA. Accelerated cellular senescence in degenerate intervertebral discs: a possible role in the pathogenesis of intervertebral disc degeneration. Arthritis Res. Ther. 2007;9:R45. doi: 10.1186/ar2198.
    1. Brosens JJ, Hayashi N, White JO. Progesterone receptor regulates decidual prolactin expression in differentiating human endometrial stromal cells. Endocrinology. 1999;140:4809–4820. doi: 10.1210/endo.140.10.7070.
    1. Mazur EC, et al. Progesterone receptor transcriptome and cistrome in decidualized human endometrial stromal cells. Endocrinology. 2015;156:2239–2253. doi: 10.1210/en.2014-1566.
    1. Christian M, et al. Interferon-gamma modulates prolactin and tissue factor expression in differentiating human endometrial stromal cells. Endocrinology. 2001;142:3142–3151. doi: 10.1210/endo.142.7.8231.
    1. Mokhtar NM, et al. Progestin regulates chemokine (C-X-C motif) ligand 14 transcript level in human endometrium. Mol. Hum. Reprod. 2010;16:170–177. doi: 10.1093/molehr/gap100.
    1. Raneros AB, et al. Increasing TIMP3 expression by hypomethylating agents diminishes soluble MICA, MICB and ULBP2 shedding in acute myeloid leukemia, facilitating NK cell-mediated immune recognition. Oncotarget. 2017;8:31959–31976. doi: 10.18632/oncotarget.16657.
    1. Altmae S, et al. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers. Sci. Rep. 2017;7:10077. doi: 10.1038/s41598-017-10098-3.
    1. Vento-Tormo R, et al. Single-cell reconstruction of the early maternal-fetal interface in humans. Nature. 2018;563:347–353. doi: 10.1038/s41586-018-0698-6.
    1. Lash GE, et al. Standardisation of uterine natural killer (uNK) cell measurements in the endometrium of women with recurrent reproductive failure. J. Reprod. Immunol. 2016;116:50–59. doi: 10.1016/j.jri.2016.04.290.
    1. Emera D, Romero R, Wagner G. The evolution of menstruation: a new model for genetic assimilation: explaining molecular origins of maternal responses to fetal invasiveness. Bioessays. 2012;34:26–35. doi: 10.1002/bies.201100099.
    1. Evans J, Salamonsen LA. Inflammation, leukocytes and menstruation. Rev. Endocr. Metab. Disord. 2012;13:277–288. doi: 10.1007/s11154-012-9223-7.
    1. Evans J, Salamonsen LA. Decidualized human endometrial stromal cells are sensors of hormone withdrawal in the menstrual inflammatory cascade. Biol. Reprod. 2014;90:14.
    1. O’Leary MA, et al. The placental mammal ancestor and the post-K-Pg radiation of placentals. Science. 2013;339:662–667. doi: 10.1126/science.1229237.
    1. Kane N, Kelly R, Saunders PT, Critchley HO. Proliferation of uterine natural killer cells is induced by human chorionic gonadotropin and mediated via the mannose receptor. Endocrinology. 2009;150:2882–2888. doi: 10.1210/en.2008-1309.
    1. Gibson DA, Greaves E, Critchley HO, Saunders PT. Estrogen-dependent regulation of human uterine natural killer cells promotes vascular remodelling via secretion of CCL2. Hum. Reprod. 2015;30:1290–1301. doi: 10.1093/humrep/dev067.
    1. Ewington LJ, Tewary S, Brosens JJ. New insights into the mechanisms underlying recurrent pregnancy loss. J. Obstet. Gynaecol. Res. 2019;45:258–265. doi: 10.1111/jog.13837.
    1. Santamaria X, Mas A, Cervello I, Taylor H, Simon C. Uterine stem cells: from basic research to advanced cell therapies. Hum. Reprod. Update. 2018;24:673–693. doi: 10.1093/humupd/dmy028.
    1. Tal R, et al. Adult bone marrow progenitors become decidual cells and contribute to embryo implantation and pregnancy. PLoS Biol. 2019;17:e3000421. doi: 10.1371/journal.pbio.3000421.
    1. Tewary, S. et al. Impact of sitagliptin on endometrial mesenchymal stem-like progenitor cells: A randomised, double-blind placebo-controlled feasibility trial. EBioMedicine, 10.1016/j.ebiom.2019.102597 (2020).
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Lun ATL, et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 2019;20:63. doi: 10.1186/s13059-019-1662-y.
    1. Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 2015;33:495–502. doi: 10.1038/nbt.3192.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4:44–57. doi: 10.1038/nprot.2008.211.
    1. Trapnell C, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 2014;32:381–386. doi: 10.1038/nbt.2859.
    1. van Dijk D, et al. Recovering gene interactions from single-cell data using data diffusion. Cell. 2018;174:716–729. doi: 10.1016/j.cell.2018.05.061.

Source: PubMed

3
S'abonner