Clearance of senescent decidual cells by uterine natural killer cells in cycling human endometrium

Paul J Brighton, Yojiro Maruyama, Katherine Fishwick, Pavle Vrljicak, Shreeya Tewary, Risa Fujihara, Joanne Muter, Emma S Lucas, Taihei Yamada, Laura Woods, Raffaella Lucciola, Yie Hou Lee, Satoru Takeda, Sascha Ott, Myriam Hemberger, Siobhan Quenby, Jan Joris Brosens, Paul J Brighton, Yojiro Maruyama, Katherine Fishwick, Pavle Vrljicak, Shreeya Tewary, Risa Fujihara, Joanne Muter, Emma S Lucas, Taihei Yamada, Laura Woods, Raffaella Lucciola, Yie Hou Lee, Satoru Takeda, Sascha Ott, Myriam Hemberger, Siobhan Quenby, Jan Joris Brosens

Abstract

In cycling human endometrium, menstruation is followed by rapid estrogen-dependent growth. Upon ovulation, progesterone and rising cellular cAMP levels activate the transcription factor Forkhead box O1 (FOXO1) in endometrial stromal cells (EnSCs), leading to cell cycle exit and differentiation into decidual cells that control embryo implantation. Here we show that FOXO1 also causes acute senescence of a subpopulation of decidualizing EnSCs in an IL-8 dependent manner. Selective depletion or enrichment of this subpopulation revealed that decidual senescence drives the transient inflammatory response associated with endometrial receptivity. Further, senescent cells prevent differentiation of endometrial mesenchymal stem cells in decidualizing cultures. As the cycle progresses, IL-15 activated uterine natural killer (uNK) cells selectively target and clear senescent decidual cells through granule exocytosis. Our findings reveal that acute decidual senescence governs endometrial rejuvenation and remodeling at embryo implantation, and suggest a critical role for uNK cells in maintaining homeostasis in cycling endometrium.

Keywords: Endometrium; Immunosurveillance; Implantation; Senescence; human; human biology; medicine; receptivity; uterine natural killer cells.

Conflict of interest statement

No competing interests declared.

Figures

Figure 1.. Decidualization induces acute senescence in…
Figure 1.. Decidualization induces acute senescence in a subpopulation of EnSCs.
(A) Representative SAβG staining in undifferentiated EnSCs (Day 0) or cells decidualized for the indicated time points with 8-bromo-cAMP and MPA. Scale bar = 100 µm. (B) SAβG activity, expressed in fluorescence intensity units (FIU), in undifferentiated EnSCs (day 0) or cells decidualized for the indicated time points. (C) Representative Western blot analysis of p53, p16, LMNB1, HMGB2, mH2A, H3K9me3 and H.H1 levels in undifferentiated EnSCs and cells decidualized for the indicated time points. β-actin served as a loading control. (D) Left panel: representative immunofluorescence staining for p16 expression in undifferentiated cells and cells decidualized for 8 days. Nuclei were counterstained with DAPI. Scale bar = 50 µm. Right panel: percentage of p16+ cells. (E) Left panel: representative confocal microscopy images of undifferentiated (Day 0) or decidualized (Day 8) EnSCs immune-probed for LMNB1, mH2A, H3K9me3 and H.H1. Scale bar = 10 µm. Right panel: nuclear size of undifferentiated EnSCs (n = 48) and of cells first decidualized for 8 days with 8-br-cAMP and MPA (C + M) (n = 48) was measured in three primary cultures. (F) Secretion of IL-8, GROα, and IL-6 was measured in the supernatant of primary EnSCs collected every 48 hr over an 8 day decidualization time-course. Data are mean ±SEM of 3 biological replicates unless stated otherwise. **p<0.01, ***p<0.001. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05.
Figure 1—figure supplement 1.. Decidualization-associated acute senescence…
Figure 1—figure supplement 1.. Decidualization-associated acute senescence in primary EnSCs.
(A) Representative SAβG staining in undifferentiated (Day 0) and decidualized (Day 8) EnSCs at the indicated passage numbers. Scale bar = 100 µm. (B) Quantitative SAβG activity at indicated passage in undifferentiated EnSCs (day 0) or cells decidualized for the specified time points (n = 6). (C) Days in culture in relation to passage number. (D) Densitometric analysis of senescence marker Western blots shown in Figure 1C. Significance for individual markers was determined by t-test in comparison to day 0. *=P < 0.05, **=P < 0.01 and ***=P < 0.001. All data are mean ±SEM of 3 biological replicates, unless indicated otherwise. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05.
Figure 2.. Senescent cells in cycling human…
Figure 2.. Senescent cells in cycling human endometrium.
(A) Left panel: representative Western blot analysis of p53, p16, LMNB1, HMGB2, mH2A, H3K9me3 and H.H1 levels in whole tissue biopsies from proliferative endometrium (PE) and secretory endometrium (SE). β-actin served as a loading control. Right panel: protein levels quantified relative to β-actin by densitometry and expressed as arbitrary units (a.u.). (B) SAβG activity, expressed in fluorescence intensity units (FIU)/mg protein, was measured in biopsies from proliferative endometrium (PE; n = 7), early-secretory (ES; n = 9), mid-secretory (MS; n = 38) and late-secretory (LS; n = 19) endometrium. (C) Immunohistochemistry demonstrating distribution of p16+ cells in the stromal compartment and luminal epithelium. Scale bars = 200 µm. (D) The abundance of p16+ cells during the luteal phase in glandular epithelium, luminal epithelium and stroma compartment was analyzed by color deconvolution using ImageJ software in 308 LH-timed endometrial biopsies (average 48 samples per time point; range: 22 to 69). The centile graphs depict the distribution of p16+ cells across the peri-implantation window in each cellular compartment. Color key is on the right. Data are mean ±SEM of 3 biological replicates unless stated otherwise. **p<0.01, ***p<0.001. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05.
Figure 3.. A FOXO1/IL-8 axis drives EnSC…
Figure 3.. A FOXO1/IL-8 axis drives EnSC differentiation and senescence.
(A) SAβG activity in EnSCs either undifferentiated, or decidualized for 8 days with 8-bromo-cAMP, MPA, or a combination. (B) Top left panel: FOXO1 mRNA levels in undifferentiated EnSCs and cells treated with 8-br-cAMP and MPA (C + M) following transfection with non-targeting (NT) or FOXO1 siRNA. Other panels: Secretion of IL-8, IL-6 and GROα was measured following FOXO1 knockdown in the supernatant of primary EnSCs every 48 hr over an 8 day decidualization time-course. (C) SAβG activity in EnSCs following transfection with NT or FOXO1 siRNA. The cultures either remain untreated or decidualized for 8 days. (D) SAβG activity in undifferentiated EnSCs treated for 8 days with increasing concentrations of recombinant IL-8 and in cells decidualized for 8 days in the presence of increasing concentrations of the CXCR2 antagonist, SB265610. (E) SAβG activity in EnSCs following transfection with IL-8 siRNA. The cultures either remain untreated or decidualized for 8 days. (F) PRL and IGFBP1 transcript levels in EnSCs following transfection with IL-8 siRNA. The cultures either remain untreated or decidualized for 8 days. (G) PRL and IGFBP1 expression in undifferentiated EnSCs, cells decidualized for 8 days, and upon withdrawal of 8-br-cAMP and MPA (C + M) for the indicated days. (H) Left panel: SAβG activity in undifferentiated EnSCs, cells decidualized for 8 days, and following withdrawal of C + M for the indicated days. Right panel: representative Western blot analysis of p53, p16, LMNB1 and HMGB2 levels in undifferentiated EnSCs, cells decidualized for 8 days, and following withdrawal of C + M for the indicated days. β-actin served as a loading control. Data are mean ±SEM of 3 biological replicates unless stated otherwise. *p<0.05, **p<0.01 and ***p<0.005. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05.
Figure 3—figure supplement 1.. EnSC differentiation and…
Figure 3—figure supplement 1.. EnSC differentiation and senescence is driven by FOXO1, IL-8 and mTOR.
(A) PRL and IGFBP1 transcript levels in EnSCs following transfection with FOXO1 siRNA. The cultures either remained untreated or were decidualized for 8 days. (B) Left panel: SAβG staining in undifferentiated EnSCs that remained untreated (control) or were incubated with recombinant IL-8 (30 μM) for 8 days. SAβG staining was also performed in parallel cultures decidualized with 8-bromo-cAMP and MPA (C + M) in the absence or presence of the CXCR2 antagonist SB265610 (10 μM). Right panel: IL-8 concentration in conditioned media from decidualized EnSCs following siRNA-mediated CXCL8 (IL-8) knockdown. (C) SAβG staining (left panel) and activity (right panel) in undifferentiated (day 0) and decidualized (day 8) EnSCs in the presence of the mTOR inhibitor rapamycin. FIU: fluorescence intensity units. (D) PRL and IGFPB1 transcripts in undifferentiated EnSCs and cells decidualized for 8 days in the presence or absence of rapamycin (100 nM). All data are mean ±SEM of 3 biological replicates. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05. Scale bars = 100 μm.
Figure 4.. Functions of senescent decidual cells.
Figure 4.. Functions of senescent decidual cells.
(A) Pearson’s correlation analysis of SAβG activity in 75 matched undifferentiated primary cultures and cultures decidualized for 8 days. (B) Representative SAβG staining in undifferentiated (Day 0) and decidualizing EnSCs (Day 8) following 4 days of pretreatment with vehicle, dasatinib (250 nM) or palbociclib (1 μM). Scale bar = 100 µm. (C) PRL and IGFBP1 mRNA expression in response to pretreatment with vehicle, dasatinib or palbociclib. The cultures then remained undifferentiated or were decidualized for 8 days. (D) IL-8, IL-6 and GROα secretion was measured every 48 hr in the supernatant of primary EnSCs decidualized for the indicated time-points following pretreatment with vehicle, dasatinib or palbociclib. (E) Colony forming unit (CFU) activity in paired EnSC cultures that either remain undifferentiated (Day 0) or were decidualized for 8 days (n = 10). (F) Left panel: representative clonogenic assays established from EnSC cultures first pretreated with vehicle, dasatinib or palbociclib and then decidualized for 8 days. Right panel: CFU activity in EnSC cultures first pretreated with vehicle, dasatinib or palbociclib and then decidualized for 8 days. Data are mean ±SEM of 3 biological replicates unless stated otherwise. *p<0.05, **p<0.01 and ***p<0.001. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05.
Figure 4—figure supplement 1.. Modulation of senescence…
Figure 4—figure supplement 1.. Modulation of senescence in EnSC cultures.
(A) Pearson’s correlation analysis of SAβG staining in 18 matched undifferentiated and decidualized EnSC cultures. (B–C) Quantitative analysis of SAβG in EnSCs following exposure to senolytics ABT-263 (B) and Dasatinib (C, left panel) for 3 days, or the CDK4/CDK6-inhibitor Palbociclib for times indicated (C, right panel). (D) Colony forming unit (CFU) efficiency of EnSC following decidualization for 8 days in the absence or presence of rapamycin (100 nM). Different letters above the error bars indicates that those groups are significantly different from each other at p<0.05. Data are mean ±SEM, n = 3.
Figure 5.. uNK cell mediated immune surveillance…
Figure 5.. uNK cell mediated immune surveillance and clearance of senescent cells.
(A) Left panel: uNK cell density in the subluminal stroma was quantified using a standardized immunohistochemistry protocol in LH timed endometrial biopsies (n = 1,997). Right panel: corresponding centile graph. Color code on the left. (B) Left panel: example of the tissue distribution of CD56+ uNK cells (brown staining) at LH + 10. Scale bar = 250 μm. Right panel: Pearson’s correlation analysis of stromal cell and uNK cell densities. A total of 80 randomly selected images from 20 biopsies were analyzed. (C) Representative images of an eosin stained primary culture decidualized for 8 days incubated for 18 hr with or without uNK cells isolated from luteal phase endometrium. Scale bar = 100 μm. (D) SAβG activity in undifferentiated or day eight decidualized EnSCs co-cultured with or without uNK cells in the presence or absence of the apoptosis inhibitor Z-VAD-FMK (Z-VAD, 10 μM) or the granzyme activity inhibitor 3,4-DCI (25 μM). (E) Secretion of IL-15 secretion was measured every 48 hr in the supernatant of primary EnSCs decidualized for the indicated time-points following pretreatment with vehicle, dasatinib (250 nM) or palbociclib (1 μM). (F) SAβG activity in undifferentiated or day eight decidualized EnSCs co-cultured with or without uNK cells in the presence or absence of an IL-15 blocking antibody (1 μg/ml). Data are mean ±SEM of 3 biological replicates unless stated otherwise. Different letters above the error bars indicate that those groups are significantly different from each other at p<0.05.
Figure 5—figure supplement 1.. uNK cell mediated…
Figure 5—figure supplement 1.. uNK cell mediated immune surveillance and clearance of senescent cells.
(A) CD56 staining on cytospin preparations of isolated uNK cells. Inset: primary antibody was omitted. (B) Real-time analysis of EnSC viability using xCELLigence in the presence or absence of uNKs following treatment with 25 μM 3,4-DCI or 10 μM Z-VAD_FMK as indicated. Data are mean ±SEM from technical replicates and representative of two further biological repeat experiments. (C) SAβG activity following exposure of EnSCs to increasing concentrations of FasL or TRAIL as indicated. Parallel co-cultures of EnSCs with uNK cells are shown for comparison. (D) SAβG activity in undifferentiated or day eight decidualized EnSCs co-cultured with or without uNK cells in the presence or absence an inhibitory NKG2D receptor antibody. Different letters above the error bars indicates that those groups are significantly different from each other at p<0.05. Data are mean ±SEM, n = 3.
Figure 6.. Dynamic inter-cycle fluctuations in uNK…
Figure 6.. Dynamic inter-cycle fluctuations in uNK cell levels.
CD56 immunohistochemistry of LH-timed endometrial biopsies obtained in three different cycles in three subjects. The day of the biopsy and the percentage of CD56+ uNK cells versus stromal cells are indicated. The color of the box indicates the percentile range of uNK when adjusted for the day of biopsy. Scale = 200 μm.
Figure 6—figure supplement 1.. uNK cells in…
Figure 6—figure supplement 1.. uNK cells in repeat biopsies.
CD56 immunochemistry of LH-timed endometrial biopsies in two consecutive cycles. Day of biopsy relative to LH surge and the percentage of CD56+ uNK cells versus stromal cells are indicated. The color of the box indicates the percentile range of uNK when adjusted for the day of biopsy. Scale = 200 μm.
Figure 7.. Schematic summary.
Figure 7.. Schematic summary.
We propose that rapid endometrial growth during the proliferative phase is important for implantation as it imparts replication stress in a subpopulation of EnSCs. Upon cell cycle exit at G0/G1, this subpopulation of stressed EnSCs do not differentiate into specialist decidual cells but undergo acute cellular senescence and secrete a host of inflammatory mediators (senescence associated secretory phenotype; SASP) involved in endometrial receptivity. In parallel, Il-15 secreted by differentiated decidual cells activates uNK cells, which then target and eliminate senescent cells through granule exocytosis. Systematic clearance of acutely senescent decidual cells by uNK cells not only remodels but also rejuvenates the endometrium at the time of embryo implantation.

References

    1. Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, Takatsu Y, Melamed J, d'Adda di Fagagna F, Bernard D, Hernando E, Gil J. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133:1006–1018. doi: 10.1016/j.cell.2008.03.038.
    1. Aird KM, Iwasaki O, Kossenkov AV, Tanizawa H, Fatkhutdinov N, Bitler BG, Le L, Alicea G, Yang TL, Johnson FB, Noma KI, Zhang R. HMGB2 orchestrates the chromatin landscape of senescence-associated secretory phenotype gene loci. The Journal of Cell Biology. 2016;215:325–334. doi: 10.1083/jcb.201608026.
    1. Al-Sabbagh M, Fusi L, Higham J, Lee Y, Lei K, Hanyaloglu AC, Lam EW, Christian M, Brosens JJ. 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. Barros F, Brosens J, Brighton P. Isolation and Primary Culture of Various Cell Types from Whole Human Endometrial Biopsies. Bio-Protocol. 2016;6:e2028. doi: 10.21769/BioProtoc.2028.
    1. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Analytical Biochemistry. 1976;72:248–254. doi: 10.1016/0003-2697(76)90527-3.
    1. Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, van Deursen JM. Senescent cells: an emerging target for diseases of ageing. Nature Reviews Drug Discovery. 2017;16:718–735. doi: 10.1038/nrd.2017.116.
    1. Chowdhury D, Lieberman J. Death by a thousand cuts: granzyme pathways of programmed cell death. Annual Review of Immunology. 2008;26:389–420. doi: 10.1146/annurev.immunol.26.021607.090404.
    1. de Keizer PL. The fountain of youth by targeting senescent cells? Trends in Molecular Medicine. 2017;23:6–17. doi: 10.1016/j.molmed.2016.11.006.
    1. de Ziegler D, Bergeron C, Cornel C, Medalie DA, Massai MR, Milgrom E, Frydman R, Bouchard P. Effects of luteal estradiol on the secretory transformation of human endometrium and plasma gonadotropins. The Journal of Clinical Endocrinology and Metabolism. 1992;74:322–331. doi: 10.1210/jcem.74.2.1730810.
    1. Demidenko ZN, Zubova SG, Bukreeva EI, Pospelov VA, Pospelova TV, Blagosklonny MV. Rapamycin decelerates cellular senescence. Cell Cycle. 2009;8:1888–1895. doi: 10.4161/cc.8.12.8606.
    1. Diep CH, Charles NJ, Gilks CB, Kalloger SE, Argenta PA, Lange CA. Progesterone receptors induce FOXO1-dependent senescence in ovarian cancer cells. Cell Cycle. 2013;12:1433–1449. doi: 10.4161/cc.24550.
    1. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith O. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. PNAS. 1995;92:9363–9367. doi: 10.1073/pnas.92.20.9363.
    1. Drury JA, Tang AW, Turner MA, Quenby S. A rapid, reliable method for uNK cell density estimation. Journal of Reproductive Immunology. 2013;97:183–185. doi: 10.1016/j.jri.2012.12.002.
    1. Erlebacher A. Immunology of the maternal-fetal interface. Annual Review of Immunology. 2013;31:387–411. doi: 10.1146/annurev-immunol-032712-100003.
    1. Evans J, Salamonsen LA, Winship A, Menkhorst E, Nie G, Gargett CE, Dimitriadis E. Fertile ground: human endometrial programming and lessons in health and disease. Nature Reviews Endocrinology. 2016;12:654–667. doi: 10.1038/nrendo.2016.116.
    1. Funayama R, Saito M, Tanobe H, Ishikawa F. Loss of linker histone H1 in cellular senescence. The Journal of Cell Biology. 2006;175:869–880. doi: 10.1083/jcb.200604005.
    1. Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocrine Reviews. 2014;35:851–905. doi: 10.1210/er.2014-1045.
    1. Groll JM, Usadi RS, Lessey BA, Lininger R, Young SL, Fritz MA. Effects of variations in serum estradiol concentrations on secretory endometrial development and function in experimentally induced cycles in normal women. Fertility and Sterility. 2009;92:2058–2061. doi: 10.1016/j.fertnstert.2009.06.018.
    1. Hanna J, Goldman-Wohl D, Hamani Y, Avraham I, Greenfield C, Natanson-Yaron S, Prus D, Cohen-Daniel L, Arnon TI, Manaster I, Gazit R, Yutkin V, Benharroch D, Porgador A, Keshet E, Yagel S, Mandelboim O. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nature Medicine. 2006;12:1065–1074. doi: 10.1038/nm1452.
    1. Hapangama DK, Kamal A, Saretzki G. Implications of telomeres and telomerase in endometrial pathology. Human Reproduction Update. 2017;23:166–187. doi: 10.1093/humupd/dmw044.
    1. Iannello A, Raulet DH. Immune surveillance of unhealthy cells by natural killer cells. Cold Spring Harbor Symposia on Quantitative Biology. 2013;78:249–257. doi: 10.1101/sqb.2013.78.020255.
    1. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, Yee H, Zender L, Lowe SW. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134:657–667. doi: 10.1016/j.cell.2008.06.049.
    1. Laws MJ, Taylor RN, Sidell N, DeMayo FJ, Lydon JP, Gutstein DE, Bagchi MK, Bagchi IC. Gap junction communication between uterine stromal cells plays a critical role in pregnancy-associated neovascularization and embryo survival. Development. 2008;135:2659–2668. doi: 10.1242/dev.019810.
    1. Lucas ES, Dyer NP, Fishwick K, Ott S, Brosens JJ. Success after failure: the role of endometrial stem cells in recurrent miscarriage. Reproduction. 2016a;152:R159–R166. doi: 10.1530/REP-16-0306.
    1. Lucas ES, Dyer NP, Murakami K, Lee YH, Chan YW, Grimaldi G, Muter J, Brighton PJ, Moore JD, Patel G, Chan JK, Takeda S, Lam EW, Quenby S, Ott S, Brosens JJ. Loss of Endometrial Plasticity in Recurrent Pregnancy Loss. Stem Cells. 2016b;34:346–356. doi: 10.1002/stem.2222.
    1. Marçais A, Cherfils-Vicini J, Viant C, Degouve S, Viel S, Fenis A, Rabilloud J, Mayol K, Tavares A, Bienvenu J, Gangloff YG, Gilson E, Vivier E, Walzer T. The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells. Nature Immunology. 2014;15:749–757. doi: 10.1038/ni.2936.
    1. Matjusaitis M, Chin G, Sarnoski EA, Stolzing A. Biomarkers to identify and isolate senescent cells. Ageing Research Reviews. 2016;29:1–12. doi: 10.1016/j.arr.2016.05.003.
    1. Mosteiro L, Pantoja C, Alcazar N, Marión RM, Chondronasiou D, Rovira M, Fernandez-Marcos PJ, Muñoz-Martin M, Blanco-Aparicio C, Pastor J, Gómez-López G, De Martino A, Blasco MA, Abad M, Serrano M. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 2016;354:aaf4445. doi: 10.1126/science.aaf4445.
    1. Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nature Reviews Molecular Cell Biology. 2014;15:482–496. doi: 10.1038/nrm3823.
    1. Murakami K, Bhandari H, Lucas ES, Takeda S, Gargett CE, Quenby S, Brosens JJ, Tan BK. Deficiency in clonogenic endometrial mesenchymal stem cells in obese women with reproductive failure--a pilot study. PLoS One. 2013;8:e82582. doi: 10.1371/journal.pone.0082582.
    1. Paria BC, Lim H, Wang XN, Liehr J, Das SK, Dey SK. Coordination of differential effects of primary estrogen and catecholestrogen on two distinct targets mediates embryo implantation in the mouse. Endocrinology. 1998;139:5235–5246. doi: 10.1210/endo.139.12.6386.
    1. Park Y, Nnamani MC, Maziarz J, Wagner GP. Cis-regulatory evolution of forkhead box O1 (FOXO1), a terminal selector gene for decidual stromal cell identity. Molecular Biology and Evolution. 2016;33:3161–3169. doi: 10.1093/molbev/msw193.
    1. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Research. 2001;29:45e–45. doi: 10.1093/nar/29.9.e45.
    1. Pohnke Y, Schneider-Merck T, Fahnenstich J, Kempf R, Christian M, Milde-Langosch K, Brosens JJ, Gellersen B. Wild-type p53 protein is up-regulated upon cyclic adenosine monophosphate-induced differentiation of human endometrial stromal cells. The Journal of Clinical Endocrinology & Metabolism. 2004;89:5233–5244. doi: 10.1210/jc.2004-0012.
    1. Rajagopalan S, Long EO. Cellular senescence induced by CD158d reprograms natural killer cells to promote vascular remodeling. PNAS. 2012;109:20596–20601. doi: 10.1073/pnas.1208248109.
    1. Ritschka B, Storer M, Mas A, Heinzmann F, Ortells MC, Morton JP, Sansom OJ, Zender L, Keyes WM. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes & Development. 2017;31:172–183. doi: 10.1101/gad.290635.116.
    1. Ruifrok AC, Johnston DA. Quantification of histochemical staining by color deconvolution. Analytical and Quantitative Cytology and Histology. 2001;23:291–299.
    1. Sadaie M, Salama R, Carroll T, Tomimatsu K, Chandra T, Young AR, Narita M, Pérez-Mancera PA, Bennett DC, Chong H, Kimura H, Narita M. Redistribution of the Lamin B1 genomic binding profile affects rearrangement of heterochromatic domains and SAHF formation during senescence. Genes & Development. 2013;27:1800–1808. doi: 10.1101/gad.217281.113.
    1. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, Braisted J, Klapa M, Currier T, Thiagarajan M, Sturn A, Snuffin M, Rezantsev A, Popov D, Ryltsov A, Kostukovich E, Borisovsky I, Liu Z, Vinsavich A, Trush V, Quackenbush J. TM4: a free, open-source system for microarray data management and analysis. BioTechniques. 2003;34:374–378.
    1. Sagiv A, Biran A, Yon M, Simon J, Lowe SW, Krizhanovsky V. Granule exocytosis mediates immune surveillance of senescent cells. Oncogene. 2013;32:1971–1977. doi: 10.1038/onc.2012.206.
    1. Sagiv A, Burton DG, Moshayev Z, Vadai E, Wensveen F, Ben-Dor S, Golani O, Polic B, Krizhanovsky V. NKG2D ligands mediate immunosurveillance of senescent cells. Aging. 2016;8:328–344. doi: 10.18632/aging.100897.
    1. Salker MS, Nautiyal J, Steel JH, Webster Z, Sućurović S, Nicou M, Singh Y, Lucas ES, Murakami K, Chan YW, James S, Abdallah Y, Christian M, Croy BA, Mulac-Jericevic B, Quenby S, Brosens JJ. 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. Smith A, Tilling K, Nelson SM, Lawlor DA. Live-Birth Rate Associated With Repeat In Vitro Fertilization Treatment Cycles. Jama. 2015;314:2654–2662. doi: 10.1001/jama.2015.17296.
    1. Takano M, Lu Z, Goto T, Fusi L, Higham J, Francis J, Withey A, Hardt J, Cloke B, Stavropoulou AV, Ishihara O, Lam EW, Unterman TG, Brosens JJ, Kim JJ. Transcriptional cross talk between the forkhead transcription factor forkhead box O1A and the progesterone receptor coordinates cell cycle regulation and differentiation in human endometrial stromal cells. Molecular Endocrinology. 2007;21:2334–2349. doi: 10.1210/me.2007-0058.
    1. Team RC. Vienna, Austria: R Foundation for Statistical Computing; 2015.
    1. van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509:439–446. doi: 10.1038/nature13193.
    1. Venables W. Ripley B. Modern Applied Statistics with S-Plus. 4th edn. Springer; 1997.
    1. Xiong S, Sharkey AM, Kennedy PR, Gardner L, Farrell LE, Chazara O, Bauer J, Hiby SE, Colucci F, Moffett A. Maternal uterine NK cell-activating receptor KIR2DS1 enhances placentation. Journal of Clinical Investigation. 2013;123:4264–4272. doi: 10.1172/JCI68991.
    1. Yuan X, Saravelos SH, Wang Q, Xu Y, Li TC, Zhou C. Endometrial thickness as a predictor of pregnancy outcomes in 10787 fresh IVF-ICSI cycles. Reproductive BioMedicine Online. 2016;33:197–205. doi: 10.1016/j.rbmo.2016.05.002.
    1. Zhang S, Lin H, Kong S, Wang S, Wang H, Wang H, Armant DR. Physiological and molecular determinants of embryo implantation. Molecular Aspects of Medicine. 2013;34:939–980. doi: 10.1016/j.mam.2012.12.011.
    1. Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, Dai HM, Ling YY, Stout MB, Pirtskhalava T, Giorgadze N, Johnson KO, Giles CB, Wren JD, Niedernhofer LJ, Robbins PD, Kirkland JL. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2016;15:428–435. doi: 10.1111/acel.12445.
    1. Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O'Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann-Stroissnigg H, Gurkar AU, Zhao J, Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD, Niedernhofer LJ, Kirkland JL. The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14:644–658. doi: 10.1111/acel.12344.

Source: PubMed

3
Suscribir