Quercetin, a senolytic, enhances endometrial stromal cell decidualization through AKT and p53 signaling and reveals the potential role of senescent stromal cells in endometriosis ================================================================================================================================================================================== * Julia G Delenko * Xiangying Xue * Prodyot K Chatterjee * Nathaniel Hyman * Andrew J Shih * Robert P Adelson * Polona Safaric-Tepes * Peter K Gregersen * Christine N Metz ## Abstract Decidualization or the differentiation of endometrial stromal cells (eSCs) into secretory decidual cells is required for successful implantation and pregnancy in menstruating species. Endometriosis, a condition accompanied by chronic pelvic pain and infertility, is observed in humans and a few species that menstruate. Several studies report that endometriosis is accompanied by significantly compromised decidualization, and this may explain some of the associated infertility. Current pharmacologics treat symptoms of endometriosis but do not alter disease progression. Sampson’s theory of retrograde menstruation is the most commonly accepted mechanism that explains the delivery of menstrual tissues to the peritoneal cavity where most endometriosis lesions develop. However, the exact etiology of endometriosis remains unknown. Recent studies support that cellular senescence, a phenotype found in shed menstrual tissues, impairs decidualization. Additionally, a senolytic agent, quercetin, has been reported to improve female fertility and reduce endometriosis in pre-clinical models. We examined the effect of quercetin on decidualization and stromal cell function using cultured eSCs isolated from menstrual effluent obtained from healthy, unaffected controls and endometriosis patients. Quercetin significantly inhibits eSC proliferation and improves decidualization of eSCs obtained from both endometriosis patients and controls. Mechanistic studies using eSCs reveal that quercetin rapidly blocks AKT, ERK1/2, and PRAS40 phosphorylation and inhibits the production of IL-6 and MMP3, prominent products of the senescence-associated secretory phenotype (SASP). Additionally, quercetin promotes both p53 phosphorylation and total p53 protein levels in eSCs and this is accompanied by increased apoptosis and the loss of larger beta-galactosidase-expressing eSCs with a senescence phenotype. This is the first study to reveal that quercetin enhances decidualization via senolytic and pro-apoptotic activities downstream of AKT signaling in controls and patients with endometriosis. These results support further investigating quercetin as a treatment for endometriosis and associated infertility. Keywords * decidualization * endometriosis * fertility * AKT signaling * p53 signaling * apoptosis * senescence ## Introduction Decidualization refers to the differentiation of fibroblast-like endometrial stromal cells (eSCs) into enlarged specialized decidual stromal cells that produce growth factors needed to create a nutrient-rich uterine environment critical for implantation and successful pregnancy 1. In mice and most other placental mammals this occurs post-implantation. By contrast, decidualization spontaneously occurs during each secretory phase of the menstrual cycle in the few species that menstruate, including humans. Numerous studies report impaired decidualization by eSCs from patients with endometriosis when compared to eSCs from healthy controls 2-5. Furthermore, recent studies describe the inhibitory effects of eSC senescence and the senescence-associated secretory phenotype (SASP) on decidualization in vitro 6 and the link between dysfunctional decidualization and reduced female fertility 7. The roles of senescence in aging, cell differentiation, and tissue injury and repair have been described 8-10. Senolytic agents have been developed to treat aging-related and other conditions by selectively clearing senescent cells (i.e., growth-arrested, viable cells that have undergone metabolic and gene expression changes) 11. Quercetin, a flavonol found in fruits, vegetables, tea, seeds, nuts, and medicinal botanicals 12, is a natural senolytic. Quercetin also exhibits anti-oxidant, anti-inflammatory, and immunomodulatory activities throughout the body, including the female reproductive tract 13; 14. Quercetin supplementation improves fecundity in young female mice 15 and increases pregnancy rates in patients with polycystic ovary syndrome (PCOS) 16. Additionally, quercetin administration significantly reduces the growth of endometrial implants in rodents 17,18, supporting its potential use in treating endometriosis. Endometriosis is a common, chronic inflammatory condition defined by the growth of endometrial-like tissues outside of the uterus, mainly in the pelvic cavity. It is characterized by chronic pelvic pain and is often accompanied by a panoply of symptoms, including dysmenorrhea, dyspareunia, dysuria, dyschezia, and infertility 19. The exact causes of endometriosis and associated infertility are not well understood. The delivery of menstrual tissues to the pelvic cavity is observed in nearly all menstruators 20 and, intriguingly, endometriosis is associated with altered endometrial tissue (shed as menstrual effluent, ME) 21-24. These findings support the postulate that aberrant endometrial tissues (or ME) delivered to the pelvic cavity via retrograde menstruation contribute to the pathogenesis of endometriosis, as described by Sampson almost 100 years ago. Our recent single-cell RNA sequencing (scRNA-Seq) analysis of fresh ME-derived tissues collected at the time of menstruation reveal the enrichment of a subset of eSCs with a senescence-like phenotype and the depletion of eSCs with a decidual phenotype in patients with endometriosis vs. healthy controls 25. These findings confirm the reported decidualization defect in eSCs in endometriosis 2-5 and emphasize the potential role of senescence in endometriosis. Pharmacological therapies for endometriosis are mainly limited to treating patient symptoms with hormones and pain relievers 26. Laparoscopic surgery to remove lesions is an alternative for more severe cases. 26 However, repeat surgeries are common and there is no clear consensus on surgery for preserving fertility. Based on prior studies supporting the therapeutic potential of quercetin in pre-clinical models of endometriosis as well as its senolytic activity and potential to enhance fertility, we used ME-derived cultured eSCs from patients with endometriosis and unaffected controls to define the effects of quercetin on eSC functions related to decidualization. ## Results ### Quercetin reduces the proliferation of endometrial stromal cells (eSCs) As described for many cell types 27-29, including uterine biopsy-derived eSCs 18, quercetin (12-50μM) reduced the relative cell numbers of ME-derived eSCs obtained from healthy controls, in a dose-dependent manner (Figure 1A). ![Figure 1.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F1) Figure 1. Quercetin inhibits eSC proliferation and enhances decidualization. **(A)** Control endometrial stromal cells (eSCs) were treated with vehicle (Veh) or quercetin (Q, 6.25-50 µM) for 72 hr and the relative cell number was determined by CyQUANT™. Each point represents data from one individual’s eSCs, with the group mean ± SD for vehicle and each Q dose. **(B-C)** Control eSCs were treated with vehicle (Veh) or quercetin (1.56-50 μM) for 4 hr prior to the addition of cAMP alone (B) or cAMP+MPA (C). After 48 hr, decidualization was analyzed by measuring IGFBP1 levels by ELISA. Each point represents data from one individual’s eSCs, as IGFBP1 (percent Veh, where 100% = Veh+cAMP alone (IGFBP1)) (B) or Veh+cAMP+MPA (IGFBP1) (C), with the group median ± interquartile range (IQR) for each Q dose. **(D)** eSCs from two control subjects were treated with vehicle (Veh) or quercetin (Q, 25µM) for 4 hr followed by vehicle or cAMP+MPA prior to immunofluorescent staining and confocal imaging; images show IGFBP1 (red), phalloidin (green), and DAPI (blue) staining (at 20x magnification). The scale bars (20 µm) are indicated. IGFBP1+ cells are indicated by yellow arrows. Images showing individual channels are in Supplementary Figure S1. **(E-G)** The effects of quercetin (Q, 25 μM) on decidualization when added 4 hr pre-cAMP±MPA (E) versus at the same time as (0 hr pre-) cAMP±MPA (F) or 20 hr post-cAMP±MPA stimulation (G). IGFBP1 levels were measured by ELISA 48 hr post cAMP±MPA stimulation. Data are shown as IGFBP1 (% Veh) where 100% = Veh+cAMP alone (IGFBP1). Each point represents data from one individual’s eSCs, with the median ± IQR shown for each group comparing Veh vs. Q. *p<0.05, **p<0.01; \***|p<0.001; \**\*|\*p<0.0001; ns=non-significant. ### Quercetin enhances decidualization of cultured eSCs The differentiation of eSCs into decidual cells in vivo by progesterone (or its stable synthetic analogue, medroxyprogesterone acetate (MPA)) and the cAMP/protein kinase A signaling pathway can be replicated in vitro using cell-permeable cAMP alone or cAMP+MPA 1. Testing a range of quercetin concentrations with eSCs obtained from healthy controls revealed that quercetin enhanced both cAMP- and cAMP+MPA-induced decidualization, as quantified by IGFBP1 production, peaking between 12.5-25 μM quercetin (Figure 1B-C, respectively). We chose the higher dose of quercetin (25 µM) for the remaining studies. IGFBP1 expression by control eSCs following cAMP+MPA-induced decidualization was confirmed by confocal microscopy (Figure 1D). IGFBP1 protein was absent without deciduogenic stimulation, and as previously described 30 not all cells express IGFBP1 following cAMP+MPA stimulation. Furthermore, quercetin increased cAMP+MPA-induced IGFBP1 expression by eSCs as determined by confocal microscopy (Figure 1D). Kinetic studies showed that when added 4 hr prior to the addition of cAMP or cAMP+MPA quercetin showed better decidualization-enhancing effects (Figure 1E) versus when added at the same time as cAMP±MPA (Figure 1F) or 20 hr post-cAMP±MPA treatment (Figure 1G). ### Endometriosis-eSCs exhibit impaired decidualization when compared to control-eSCs As previously reported 5, eSCs from endometriosis cases exhibit significantly reduced decidualization following cAMP alone (Figure 2A) and cAMP+MPA (Figure 2B) when compared to eSCs obtained from controls, regardless of the decidualization stimulus. Note that eSCs from both cases and controls exhibit a wide range of decidualization responses (Figure 2). ![Figure 2.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F2) Figure 2. Endometriosis eSCs exhibit impaired decidualization. **(A-B)** Comparison of decidualization responses of eSCs from controls (CTRL) vs. endometriosis (ENDO) cases induced by cAMP alone (A) and cAMP+MPA (B). Decidualization was determined by IGFBP1 levels measured 48 hr post-cAMP±MPA by ELISA. Each point represents data from one individual’s eSCs, with mean ± SD shown for each group. **p<0.01 CTRL vs. ENDO. ### Quercetin promotes decidualization of eSCs obtained from controls and endometriosis cases Using ME-derived eSCs isolated from healthy controls and endometriosis cases, quercetin treatment significantly enhanced both cAMP-induced (Figure 3A-B) and cAMP+MPA-induced decidualization (determined by IGFBP1 levels) (Figure 3C-D) regardless of whether the donor was affected by endometriosis or not. Also, quercetin treatment of a subset of control-eSCs and endometriosis-eSCs significantly increased cAMP-induced and cAMP+MPA-induced PRL, another biomarker of decidualization (Figure 3E-H). Similar results were obtained for *IGFBP1* and *PRL* mRNA expression when analyzed by RT-qPCR (Supplementary Figure S2). ![Figure 3.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F3) Figure 3. Quercetin enhances decidualization by control-eSCs and endometriosis-eSCs, as determined by IGFBP1 and PRL production. **(A-H)** Endometrial stromal cells (eSCs) from controls (A, C, E, and G, CTRL) and endometriosis cases (B, D, F, and H, ENDO) were treated with vehicle (Veh) or quercetin (Q, 25μM) for 4 hr prior to the addition of cAMP (A-B, E-F) or cAMP+MPA (C-D, G-H). After 48 hr, decidualization was analyzed by measuring IGFBP1 (A-D) or PRL (E-H) levels by ELISA. Data points connected by a line represent paired data points from one individual’s eSCs (± Q). *p<0.05; **p<0.01; \***|p<0.001; \**\*|\*p<0.0001 comparing Veh vs. Q-treated. It has been proposed that prostaglandin E2 (PGE2) and progesterone are the minimal ancestral deciduogenic signals that induce decidualization in placental mammals 31. As shown for cAMP- and cAMP+MPA-induced decidualization (Figure 3A-H), quercetin significantly elevates both PGE2- and PGE2+MPA-induced decidualization by control-eSCs (Supplementary Figure S3). ### Quercetin does not generate intracellular cAMP production by eSCs Because intracellular cAMP ([cAMP]i) drives decidualization, and quercetin enhances [cAMP]i concentrations in the N1E-115 neuroblastoma cell line 32, we assessed the production of [cAMP]i by quercetin-treated eSCs. This approach was supported by scRNA-Seq analyses revealing the expression of multiple *ADCY* transcripts (that encode adenylyl cyclases required to synthesize 3’,5’-cAMP) by eSCs in fresh ME, including low expression of *ADCY1, 3, 6, 7,* and *9* transcripts (Supplementary Figure S4). Forskolin, a potent activator of adenylyl cyclase, significantly induced the production of [cAMP]i by control-eSCs (Figure 4A). However, quercetin treatment did not induce [cAMP]i production by control- or endometriosis-eSCs (Figure 4A-B). ![Figure 4.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F4) Figure 4. Quercetin does not increase [cAMP]i concentrations in eSCs. (**A-B**) Endometrial stromal cells (eSCs) from controls (CTRL) or endometriosis cases (ENDO) were treated with IBMX (0.1mM, a phosphodiesterase inhibitor that blocks [cAMP]i degradation), followed by addition of either vehicle, forskolin (FOR, 25µM, an activator of adenylyl cyclases and hence, [cAMP]i) or quercetin (Q, 25µM). Lysates were analyzed by ELISA for [cAMP]i concentrations (A-B). For clarity, data without forskolin (FOR) treatment are also shown on a different scale (B). Each point represents data from one individual’s eSCs, with median ± IQR shown for each group. **p<0.001 vs. vehicle (IBMX alone); ns=not significant. ### Quercetin does not exert anti-oxidant effects in eSCs Measuring oxidative stress using the DCF-DA (2’-7’-Dichlorofluorescein diacetate) assay, H2O2-treatment significantly induced oxidative stress in both control-eSCs and endometriosis-eSCs (Figure 5A-D). While the well-known antioxidant, *N*-acetylcysteine (NAC), significantly reduced H2O2-induced oxidative stress by eSCs (Figure 5A-D), quercetin did not reduce oxidative stress when added before (Figure 5A-B) or after H2O2 stimulation (Figure 5C-D). ![Figure 5.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F5.medium.gif) [Figure 5.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F5) Figure 5. Quercetin does not reduce oxidative stress in eSCs. **(A-D)** Endometrial stromal cells (eSCs) from controls (CTRL, A and C) or endometriosis cases (ENDO, B and D) were treated with either vehicle (Veh), quercetin (Q, 25μM), or *N*-acetylcysteine (NAC, 10mM) before H2O2 (500µM) (A and B) or after H2O2 (500µM) (C and D), and oxidative stress was measured 3 hr later using the DCF-DA assay. Data are shown as % Veh control (oxidative stress) where 100% = Veh-Veh-treated eSCs. Each point represents data from one individual’s eSCs, with median ± IQR shown for each group. *p<0.05; **p<0.01; \***|p<0.001; ns = not significant. ### Quercetin treatment of eSCs alters numerous signaling pathways Phospho-kinase arrays were used to investigate various signaling pathways altered by quercetin. Compared to vehicle treatment of control-eSCs, quercetin consistently reduced phospho-AKT (T308 and S473), phospho-ERK1/2 (multi), phospho-PRAS40 (T246), and phospho-WNK1 (T60) levels, and significantly upregulated phospho-p53 (S46) levels, when assessed 4 hr after treatment (Figure 6A-B, Supplementary Figure S5). ![](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F6/graphic-6.medium.gif) [](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F6/graphic-6) ![](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F6/graphic-7.medium.gif) [](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F6/graphic-7) Figure 6. Quercetin reduces AKT and ERK1/2 phosphorylation and signaling and promotes p53 (Ser46) phosphorylation. **(A-B)** Control eSCs were treated with vehicle (Veh) or quercetin (Q, 25μM) for 4 hr before analyzing cell lysates using a phospho-kinase array panel. Representative arrays (A) and quantification of array analytes/spots (B) are shown. Data for differentially expressed targets are presented as specific spot density normalized to the reference spot comparing Veh-vs. Q-treated (B). Each point represents the specific density of each duplicate analyte spot as a percentage of Veh-treated, with group median ± IQR shown for control- and endometriosis-eSCs. **p<0.01. The remaining arrays are in Supplementary Figure S5. **(C-D)** Control (CTRL) and endometriosis (ENDO) eSCs were treated with Veh or Q (25µM) for 4 hr before western blotting for p-AKT, total AKT, p-ERK1/2, total ERK1/2, p-PRAS40, and total PRAS40. Representative blots and quantification of specific analytes are shown in (C) and (D), respectively. Band densities were normalized to GAPDH and shown as fold-change between Veh-vs. Q-treated eSCs in (D). Each point represents data from one individual’s eSCs, with median ± IQR shown for each group. *p<0.05; \***|p<0.001; \**\*|\*p<0.0001. (**E)** Control-eSCs were treated with Veh or Q (25µM) for 0.5, 2, and 4 hr before western blotting for phospho-AKT, total AKT, phospho-ERK1/2, total ERK1, phospho-PRAS40, and total PRAS40. **(F-I)** Control-eSCs were treated with vehicle (Veh), AKT inhibitor MK-2206 (MK, 1μM), quercetin (Q, 25μM) or Q+MK for 4 hr prior to cAMP (F and H) or cAMP+MPA (G and I) stimulation; decidualization was assessed by measuring IGFBP1 (F and G) or PRL (H and I) by ELISA. Data are presented as fold-change in IGFBP1 or PRL over Veh-treated (Veh-treated=1). Each symbol represents data from one individual’s eSCs, with median ± IQR shown for each group. *p<0.05; **p<0.01; \***|p<0.001; ns=non-significant. We confirmed that quercetin significantly reduced the levels of phospho-AKT (S473), phospho-ERK1/2 (multi), and phospho-PRAS40 (T246) when analyzed by western blotting 4 hr later using control-eSCs and endometriosis-eSCs without changing total unphosphorylated target protein levels (Figure 6C-D). Time course analyses reveal that quercetin reduced phospho-AKT (S473), phospho-ERK1/2 (multi), and phospho-PRAS40 (T246) levels in control-eSCs within 30 min and this reduction persisted for 4 hr (Fig. 6E). Accordingly, MK-2206, the highly selective inhibitor of AKT1/2/3, significantly enhanced cAMP-induced IGFBP1 and PRL production (Figure 6F and 6H, respectively) and cAMP+MPA-induced IGFBP1 and PRL production (Figure 6G and 6I, respectively). The effect of MK-2206 on decidualization was similar to that achieved by quercetin and no additive effects were observed when MK-2206 was combined with quercetin (Figure 6F-I), supporting a similar or overlapping pathway. ### Quercetin increases p53 expression Although quercetin-induced phospho-p53 (Ser46) levels when analyzed using the phospho-kinase array (Figure 6A-B), phospho-p53 (Ser46) was not detected by western blotting methods due to lower detection sensitivity when compared to the array’s capture method (data not shown). However, quercetin significantly increased total p53 protein levels by eSCs after 4 hr (Figure 7A-B). Although higher p53 levels were not detected after 2 days post-quercetin (Figure 7C-D), the increase in total p53 protein was detected as early as 2 hr post-quercetin (Figure 7E). Consistent with these observations, pifithrin (PFT, 40µM), an inhibitor of p53, significantly reduced cAMP+MPA-decidualization when compared to vehicle (Figure 7F) and caused a similar reduction in quercetin-enhanced cAMP+MPA-decidualization (Figure 7G). By contrast, nutlin-3a, an inhibitor of MDM2 and promoter of p53 stability, partially improved decidualization when added post-cAMP+MPA (Figure 7H). ![Figure 7.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F7.medium.gif) [Figure 7.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F7) Figure 7. Quercetin induces p53, which contributes to decidualization. **(A-D)** Control (CTRL)- and endometriosis (ENDO)-eSCs were treated with vehicle or quercetin (Q, 25 µM) for 4 hr (A, B) or 2 days (C, D) before western blotting for p53 and GAPDH levels. Representative blots are shown in (A) and (C). Band densities for control-eSCs were normalized to GAPDH and presented as fold-change after 4 hr (B) or 2 days (D) following quercetin treatment, with median ± IQR shown for each group. \***|p<0.001 comparing fold-change in GAPDH vs. p53. **(E)** Control-eSCs were treated with Veh or Q (25 µM) for 0.5, 2, and 4 hr before western blotting for p53 and GAPDH. (**F-G)** Control-eSCs were treated with vehicle or Pifithrin α alone (PFT, 40 μM) for 4 hr prior to cAMP+MPA stimulation (F) or PFT (40 μM) and Q (25μM) for 4 hr prior to cAMP+MPA stimulation (G). Decidualization was assessed by measuring IGFBP1 levels by ELISA post-cAMP+MPA. **(H)** Control-eSCs were treated with Veh or nutlin-3a (Nut, 100nM) 24 hr after cAMP+MPA stimulation and decidualization was assessed by measuring IGFBP1 production 48 hr post-cAMP+MPA. For F-H, data are presented as IGFBP1 percent vehicle (Veh-treated cAMP+MPA=100%). Each point represents data from one individual’s eSCs, with median ± IQR shown for each group. **p<0.01 vs. Veh-treatment (F and H) or Q alone (G). ### eSCs show evidence of a senescence-like phenotype, which is reduced by quercetin treatment Our recent studies revealed numerous senescence-related gene markers that were consistently upregulated in eSCs of fresh ME tissues obtained from endometriosis patients vs. controls 25. Under basal conditions, early passage eSCs (p2-p3) express SASP biomarkers IL-6 and MMP3 (Figure 8A), and this SASP phenotype is reduced by quercetin treatment (Figure 8A-C). Also under basal conditions, early passage control-eSCs (p2-p3) express senescence-related markers p21 and p16 (which increase with senescence), and lamin B1 (which decreases with senescence) (Figure 8D). As expected, treatment with 250 µM H2O2, previously shown to induce senescence in eSCs 33, further induced p16, p21, and MMP3 levels and reduced lamin B1 levels over time (Figure 8D). Also, H2O2 treatment reduced decidualization (Figure 8E), while quercetin significantly restored this inhibition of decidualization (Figure E). ![Figure 8.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F8.medium.gif) [Figure 8.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F8) Figure 8. eSCs show evidence of senescence phenotype, which is reversed by quercetin. **(A)** Endometriosis (ENDO)-eSCs and control (CTRL)-eSCs were treated with vehicle or quercetin (Q, 25 µM) for 2 days before western blotting for SASPs, IL-6 and MMP3, or GAPDH. A representative blot is shown. **(B-C)** Quantification of blots (with band densities normalized to GAPDH) from control-eSCs treated with vehicle (Veh) or quercetin (Q, 25 µM) for 2 days for IL-6 and MMP3 is shown in (B) and (C), respectively. Data are presented as IL-6 percent vehicle or MMP3 percent vehicle (where Veh-treated=100%). Each point represents data from one individual’s eSCs, with median ± IQR shown for each group *p<0.05; **p<0.01; ns=non-significant. **(D)** Control-eSCs were treated with vehicle or 250 µM H2O2 for 2 hr and then, eSCs were harvested at 2, 4, 6, and 8 days post-H2O2 exposure for western blotting analysis for p21, p16, lamin B1, MMP3, and GAPDH. **(E)** Control-eSCs were treated with vehicle (Veh) or H2O2 (250 µM) for 2 hr and then treated with vehicle (Veh) or quercetin (Q, 25 µM), as indicated, for 4 hr prior to cAMP+MPA-induced decidualization. After 48 hr, culture supernatants were assessed for IGFBP1 (pg/ml) by ELISA. Data points connected by lines represent paired data points from one individual’s eSCs (comparing Veh vs H2O2 and H2O2-Q vs. H2O2+Q). *p<0.05; **p<0.01 Veh vs. Q-treated; ns=non-significant. ### Inhibition of apoptosis blocks decidualization; quercetin induces apoptosis in a subset of eSCs Based on the role of p53 (S46) in apoptosis and the increase in p-p53 (Ser46) and p53 expression by quercetin, we tested the effect of Z-VAD-fmk, a pan-caspase inhibitor that prevents apoptosis, on decidualization. Pretreating control-eSCs with Z-VAD-fmk (40 µM) significantly reduced decidualization and blocked quercetin-enhanced decidualization when compared to vehicle-treated eSCs (Figure 9A-B). Accordingly, quercetin significantly increased eSC apoptosis in a subset of eSCs when analyzed by Annexin V staining by flow cytometry 24 hr and 48 hr later (Figure 9C-D). In addition, using a flow cytometry-based method with a senescence-associated beta-galactosidase (SA-βgal) substrate to detect senescent cells we found that quercetin treatment significantly decreased the fraction of larger SA-βgal+/senescent cells (Figure 9E-F). ![Figure 9.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F9.medium.gif) [Figure 9.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F9) Figure 9. Inhibiting apoptosis blocks decidualization and quercetin induces apoptosis in a subset of eSCs. **(A-B)** Control-eSCs (p3) were treated with vehicle (Veh) or Z-VAD-fmk (0 vs. 40 µM), a pan-caspase inhibitor that blocks apoptosis, in the presence of vehicle (A) or quercetin (Q, 25 µM) (B) prior to inducing decidualization with cAMP+MPA. Decidualization was assessed 48 hr later by ELISA for IGFBP1. Each point represents data from one individual’s eSCs, with the median ± IQR shown for each group. **p<0.01. (**C-D**) Quercetin induces apoptosis in a subset of control-eSCs (passage 3-4), as determined by flow cytometry. Representative gating (upper panels) and histograms (lower panels) of Annexin V staining comparing vehicle-treated (Veh, left panels) vs. quercetin-treated (Q, 25 µM, right panels) eSCs 24 hr post-treatment (C). Quantification of quercetin-induced apoptosis (shown as fold-change) measured 24 hr and 48 hr post-vehicle (Veh) vs. post-quercetin (Q, 25 µM) treatment (D). Each point represents data from one individual’s eSCs (fold-change in apoptosis), with median ± IQR shown for each group (±Q). **p<0.01. (**E-F**) Larger senescence-associated β-galactosidase (SA-βgal)+ control-eSCs are reduced following quercetin treatment. SA-βgal was determined using 9H-(1,3-Dichloro-9,9-Dimethylacridin-2-one) β-D-Galactopyranoside (DDAOG), a substrate of SA-βgal that yields a far-red fluorescent product detected by flow cytometry at 670nm. Y-axis (FSC-A, size) and X-axis (DDAOG, 670nm). Representative flow cytometry plots show the gating of larger DDAOG, 670nm/SA-βgal (Beta-gal)+ eSCs comparing vehicle-treated (Veh – left panel) vs. quercetin-treated (Q, 25 µM-right panel) eSCs (p3-4) at 48 hr post-treatment (E). The percentage of Beta-gal+/senescent cells (of total viable cells per eSC sample) is indicated on the representative plots in E. The fold-change in the number of larger Beta-gal+/senescent cells comparing vehicle (Veh) vs. quercetin (Q, 25 µM) is shown (F). Each point represents data from one individual’s eSCs (fold-change in larger Beta-gal+ cells), with median ± IQR shown for each group (±Q). *p<0.05 comparing vehicle vs. Q. ## Discussion Here, we report that quercetin, a natural senolytic agent, decreases proliferation and increases decidualization of ME-derived eSCs. For the first time we show that quercetin significantly enhances decidualization by eSCs obtained from both healthy controls and endometriosis cases (which generally show impaired decidualization responses) using cAMP alone and cAMP+MPA as deciduogenic stimuli. Previous studies have linked quercetin’s effect on decidualization to its anti-proliferative effect 17; 18. However, not all agents that inhibit eSC proliferation enhance decidualization responses, suggesting that effects are likely mediated through specific signaling pathways. We show that quercetin rapidly reduces the phosphorylation of AKT (S473/T308), as well as ERK1/2 (multi), PRAS40 (T246), and WNK1 (T60) and increases the phosphorylation of p53 (S46) and the expression of total p53 protein. Additionally, we reveal a senescence phenotype in low passage eSCs similar to that observed in a subset of eSCs in fresh ME 25 (which are delivered to the pelvic cavity during menstruation). Finally, our data support the induction of apoptosis by quercetin, likely in senescent cells, leading to enhanced decidualization. While prior studies report that p53 (S46) is firmly linked to apoptosis and that quercetin induces apoptosis, these findings have not been shown with eSCs nor have they been connected to the decidualization response and female fertility. These studies are the first in-depth exploration of the effect of the senolytic, quercetin, on decidualization to reveal its regulation of downstream AKT signaling and p53 levels and to relate these findings to the delivery of senescent ME-derived eSCs to the pelvic cavity during menstruation (or retrograde menstruation) highlighting the role of senescence in endometriosis. It is well-established that progesterone raises [cAMP]i levels to trigger decidualization in vivo, leading to the differentiation of fibroblast-like eSCs into decidual cells, and this process can be recapitulated with cultured eSCs using cell-permeable cAMP ± MPA 1. Using this model system, numerous studies report decidualization defects with uterine biopsy-derived eSCs 2; 3; 34 and ME-derived eSCs 4; 5 in the setting of endometriosis. Here, we report that quercetin enhanced the decidualization response regardless of whether eSCs were obtained from endometriosis cases or unaffected healthy controls (Figure 3). However, quercetin did not promote [cAMP]i levels (Figure 4), nor did it appear to mediate decidualization through anti-oxidant activity (Figure 5). Consistent with prior studies revealing AKT dephosphorylation during decidualization and increased phosphorylation of AKT levels (i.e. increased AKT activation) by stromal cells in the eutopic endometrium of endometriosis cases and endometriosis lesions 35-37, quercetin reduced AKT phosphorylation in eSCs. We propose that persistent phospho-AKT activity may explain decidualization defects observed in the setting of endometriosis which can be corrected with quercetin – as quercetin rapidly reduced AKT phosphorylation in eSCs (within 30 min, Figure 6E). Likewise, the AKT inhibitor, MK-2206, significantly increased decidualization (Figure 6F-G). AKT is a pro-survival factor with potent apoptosis-inhibiting activity whose function in regulating transcription factors and other proteins is mediated, in part, by its phosphorylation status. Thus, our findings are consistent with the reported ‘AKT inhibitory action’ of quercetin on eSCs 18 and breast cancer cells 38 and the association between inhibition of AKT signaling and increased decidualization and fertility 39;40. AKT phosphorylates numerous proteins, including ‘with no lysine kinase-1’ or WNK1 (T60) 41 and ‘proline-rich AKT substrate of 40 kDa’ (PRAS40), which in turn enhances activation of PI3K/AKT signaling 42. Accordingly, we found that quercetin reduced the phosphorylation of WNK1 and PRAS40 by eSCs within 30 min (Figure 6E). Interestingly, PRAS40 plays important roles in cellular senescence and p53 regulation 43. Specifically, phospho-PRAS40 decreases p53 expression level 43 and inhibits pro-apoptotic gene expression 44. Our data suggest that quercetin signals through this pathway to enhance p53 stability. Additionally, we found that quercetin-treated eSCs show a reduction in the phosphorylation of ERK1/2 (Figure 6A-D). ERK1/2 regulates a parallel signaling pathway to the AKT pathway and downstream of receptor tyrosine kinase activation. It regulates proliferation, differentiation, cell survival and apoptosis, and negatively regulates p53. See Figure 10. ![Figure 10.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F10.medium.gif) [Figure 10.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F10) Figure 10. The AKT and ERK1/2 signaling pathways promote endometrial stromal cell survival and proliferation and suppress apoptosis. Quercetin (QUE) promotes apoptosis in eSCs by inhibiting the AKT and ERK1/2 pathways and enhancing p53 stability leading to the apoptosis of potentially senescent cells. Together, these QUE activated pathways limit cell proliferation and survival and may eliminate senescent cells and reduce the progression of endometriosis. Solid black lines represent known pathways. Green and red solid lines indicate positive and negative effects of quercetin, respectively, based on our data. Dashed green and dashed red lines indicate proposed positive and negative effects of QUE, respectively. Together, our data with published data support the role of senescence in endometriosis (dashed black lines). Created with [BioRender.com](http://BioRender.com). Among the phospho-kinase targets analyzed, only p53 (S46) showed increased phosphorylation by quercetin (Figure 6A-B). Additionally, quercetin increased total p53 protein levels (Figure 7). Our findings differ from a previous report that quercetin enhances decidualization by decreasing *TP53* gene expression 45. We did not explore *TP53* mRNA expression but based on our findings, we propose that quercetin likely enhances p53 stability. While poorly understood, the role of p53 in female fertility has been reported 46-49. Female *TP53*-knockout mice have smaller litter sizes and decreased fertility compared to wild-type mice 50. Like most mammals, rodents do not menstruate and decidualize after implantation. By contrast, the few species that menstruate, including humans, spontaneously decidualize prior to implantation51; 52. It is important to note that the Arg/Arg and Arg/Pro72 variants of the *TP53* gene are potent inducers of apoptosis, while the Pro/Pro72 variant of *TP53* impairs apoptosis 53; 54. Interestingly, patients carrying the Pro/Pro72-*TP53* gene are overrepresented among patients with recurrent implantation failure and patients undergoing in vitro fertilization (IVF) 55; 56. In fact, the implantation rate in IVF patients carrying the Pro/Pro72-*TP53* genotype is approximately one-half the rate of Arg/Arg72-*TP53* carriers (approximately 20% vs. 40%, respectively) 55; 57; 58. Correspondingly, their pregnancy rates are reduced compared to Arg/Arg72 carriers 55; 57; 58. Together with our data here, it is reasonable to propose that quercetin induces p53 (S46) phosphorylation to promote apoptosis in a subset of senescent eSCs that likely exert inhibitory effects on decidualization. This postulate may not be testable using most animal models because rodents and other non-menstruating species express a p53 protein lacking the S46 residue implicated in apoptosis 59. By contrast, eSCs from humans and other menstruating mammals express p53 (S46) known to mediate apoptosis 59. Recent studies suggest that senescent eSCs inhibit decidualization 6; 60. Also, our prior scRNA-Seq data revealed that a subset of eSCs with a senescent phenotype in ME are enhanced among endometriosis cases when compared to unaffected controls 25. Senescent cells in ME tissues delivered to the pelvic cavity via retrograde menstruation may promote endometriosis. Senescent cells are growth arrested, secrete SASP-related inflammatory mediators and reactive oxygen species (ROS), and mostly resist apoptosis 61; 62. As a polypharmacologic agent, quercetin exerts both anti-inflammatory and anti-oxidant effects 63; 64 and overcomes resistance to apoptosis in vitro and in vivo 65-68. Here, quercetin’s anti-oxidant effects did not appear to play a role in improving decidualization (Figure 5). Nor did quercetin increase intracellular cAMP (Figure 4). Interestingly, we found that at baseline low passage (passage <3) ME-derived eSCs express several senescence-related markers (e.g., p21, p16 and lamin B1), as well as SASP-related IL-6 and MMP3 (Figure 8A-B) – similar to markers found in fresh ME 25. Also, the expression of these senescence markers was further induced by H2O2, and both their baseline and H2O2-induced expression were blocked by quercetin (Figure 8). Inhibiting apoptosis with Z-VAD-fmk, a pan-caspase inhibitor, reduced decidualization and limited the enhancing effect of quercetin on decidualization (Figure 9). Consistent with quercetin’s ability to induce apoptosis in non-tumor cells 65 and senescent cells 66, quercetin increased apoptosis in a subset of eSCs that are likely senescent, as shown by the loss of SAβ-gal-expressing eSCs following quercetin (Figure 9). While the hallmark of senescence is irreversible loss of cellular proliferation, there are no additional global characteristic feature or phenotype for all senescent cells. The features of senescent cells vary depending cell type, differentiation status, microenvironment, senescence stimulus, and other factors. Therefore, there are major challenges to identifying and studying senescent cells. While we show that low passage eSCs express several senescence-like features and quercetin induces apoptosis of eSCs with a concomitant loss of large SAβ-gal+ eSCs, additional studies are needed to confirm that quercetin preferentially induces apoptosis in senescent cells. Our findings highlight the importance of studying ME-derived eSCs to better understand their potential role in the abdominal cavity where ME-tissues are delivered each month during menstruation and where most endometriosis lesions occur. Additionally, since endometriosis and adenomyosis share similar pathophysiology 69, trapped senescent eSCs in the endometrium may contribute to adenomyosis, a condition where endometrial cells invade the uterine wall. Interestingly, the prevalence of adenomyosis in a recent study of infertile patients with endometriosis was 79% 70. Quercetin has been used as a senolytic agent in many clinical trials and pre-clinical trials for numerous conditions 71. Additionally, it has been proposed to increase female fertility. Specifically, quercetin increases fecundity in rodents 15, and in a small randomized clinical trial of patients with PCOS quercetin treatment led to improved hormonal regulation and pregnancy rates 16. Additionally, quercetin when combined with other agents, including curcumin, reduced endometriosis-associated pain in a small cohort 72. Our results are consistent with prior studies using murine models that reported the beneficial effects of quercetin in endometriosis, in part by limiting cell proliferation 17; 18. Furthermore, we reveal a senescence phenotype in low passage ME-derived eSCs, consistent with prior studies with fresh ME 25 and show that quercetin improves decidualization via specific signaling pathways implicated in regulating fertility (AKT and p53) and apoptosis. Together, these findings warrant future studies to explore the role of senescence in the pathogenesis of endometriosis and the use of quercetin and other senolytic agents as potential therapies or adjunct treatments for endometriosis and associated infertility. ## Material and methods ### Materials Quercetin was purchased from Alfa Aesar/Fisher Scientific (Waltham, MA, US). 8-bromoadenosine 3′,5′-cyclic monophosphate sodium salt (cAMP), IBMX (3-isobutyl-1-methylxanthine), forskolin, hydrogen peroxide (H2O2), medroxyprogesterone acetate (MPA), and *N*-acetylcysteine (NAC) were purchased from Sigma-Aldrich (St. Louis, MO, US). Prostaglandin E2 (PGE2), Z-VAD-fmk, pifithrin-α (PFT), nutlin-3a (Nut), and MK-2206 were purchased from MedChemExpress, LLC (Monmouth Junction, NJ, US). ### Isolation and culture of menstrual effluent (ME)-derived endometrial stromal cells This study was conducted with approval from the Institutional Review Board (IRB) of Northwell Health (IRB #13-376A). Women of reproductive age (18 to 45 years old) who were not pregnant or breastfeeding or on hormonal contraceptive, and who were menstruating and willing to provide menstrual effluent (ME) samples were consented. Patients with histologically confirmed endometriosis (documented in a pathology report following laparoscopic surgery and excision of lesions) were recruited and enrolled as ‘endometriosis’ participants (cases) through the ROSE study. Control subjects (controls) who self-reported no history suggestive of a diagnosis of endometriosis were also recruited and enrolled through the ROSE study. No subjects reported having polycystic ovary syndrome (PCOS) or adenomyosis. Endometrial stromal cells (eSCs) from ME collected from cases and controls were grown, as previously described 5. Briefly, ME-derived eSCs were grown in DMEM containing 10% MSC-fetal bovine serum (FBS), 1% penicillin-streptomycin-glutamine (PSQ) (maintenance media) (Gibco/Thermo-Fisher), and normocin (1:500) (Invivogen, San Diego, CA, US) (maintenance media) at 37°C with 5% CO2; cells were split 1:6. Passage 1-3 eSCs were used for experiments (unless otherwise indicated) or were cryopreserved in 10% DMSO/90% FBS for later use. Formal sample size calculations were not performed, as the study of ME-derived stromal cells does not involve ethical, time or cost issues which warrant sample size calculations 73. Sample sizes for functional cell-based assays were consistent with our prior studies 4; 5. ### eSC proliferation assays ME-derived eSCs were plated in 96-well plates (100 µL/well, 1.5X104/mL) in maintenance media. The next day, media was aspirated and replaced with DMEM containing 2% FBS+1% PSQ and normocin (1:500) (assay media) and treated with vehicle or quercetin (0-50µM) (n=4-6 replicates per condition) at 37°C with 5% CO. After a 72 hr incubation, cells were washed once with cold PBS, aspirated, and frozen at −80°C until assayed for cell proliferation using the CyQUANT™ Cell Proliferation Assay, according to the manufacturer’s directions (Thermo-Fisher, Waltham, MA, US). CyQUANT™ dye binds to DNA, and the fluorescence emitted by the dye is linearly proportional to the number of cells in the well. Data are shown as relative cell number for each subject’s cells, with a group mean±SD (for normally distributed data) indicated for quercetin concentrations (0-50µM). ### Decidualization assays using ME-derived cultured eSCs In the morning, confluent monolayers of eSCs (passages 1-3) isolated/grown from controls and endometriosis cases were lifted with trypsin/EDTA, washed, resuspended in maintenance media (DMEM, 10% FBS, PSQ, and normocin), and plated in 96-well plates (100 µL/well, 2.5X105/mL). The numbers of subjects are shown in each figure. The same evening, media was aspirated and replaced with decidualization assay media (DMEM, 2% FBS, PSQ, and normocin). After an overnight incubation, cells were treated with either vehicle or quercetin (25μM) for 4 hr (unless described otherwise) before the addition of cAMP (0.5 mM) + MPA (10-7 M) to induce decidualization (assays were performed in duplicates or triplicates). The dose and timing of quercetin addition were based on the literature and optimization studies using eSCs obtained from control subjects. After 48 hr (following cAMP+MPA treatment), supernatants were collected and frozen at −80°C until assayed in duplicate or triplicate for IGFBP1 or PRL proteins by ELISA (R&D Systems, Minneapolis, MN), as previously described 5. Protein data for decidualization assays are shown as IGFBP1 or PRL protein (pg/ml) or percent control (vehicle-treated=100%) found in cell-free culture supernatants after 48 hr, unless otherwise specified. In a subset of control and endometriosis subjects’ cells, decidualization was analyzed by measuring *IGFBP1* and *PRL* mRNA expression by quantitative reverse transcription PCR (qRT-PCR). Briefly, high quality RNA was isolated from cells using the RNeasy Universal Plus Mini Kit (Qiagen). RNA quality was analyzed using the NanoDrop spectrophotometer (Wilmington, DE, US) and the Bioanalyzer (Agilent Technologies Genomics, Santa Clara, CA, US); samples with OD260:280 and OD260:230 ratios >1.9 were converted to double stranded cDNA using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, US). Real time qPCR (RT-qPCR) reactions using specific primers and the Roche Universal Probe Library (see Supplementary Table S1) were performed in triplicate using the Eurogentec qPCR MasterMix Plus (AnaSpec, Inc, Fremont, CA, US) and the Roche LightCycler 480. Relative changes in gene expression were calculated as fold-changes using the comparative 2^(-ΔΔCt) method; based on optimization studies, human *HPRT1* was used as the housekeeping gene for normalizing transcript levels, as previously described 5. The effects of MK-2206 (AKT inhibitor), pifithrin-α (p53 inhibitor), nutlin-3a (p53 activator), and Z-VAD (pan-caspase inhibitor/apoptosis inhibitor) on decidualization were examined as described above, except vehicle, MK-2206 (1μM), Z-VAD (40μM) or PFT (40μM) was added to eSC cultures with vehicle or quercetin (25μM); 4 hr later cells were treated with cAMP alone or cAMP±MPA and processed as described above. Nutlin-3a (100nM) was added 24 hr following cAMP+MPA treatment. Doses were chosen based on the literature and results of in-house optimization studies with eSCs where cytotoxicity (assessed by neutral red 74) was not detected. The numbers of subjects are shown in each figure and for each subject both decidualization assays and ELISA assays were performed in duplicate or triplicate. ### Immunofluorescent staining for IGFBP1 and assessment by confocal microscopy Two control subjects’ eSCs were grown to confluence on glass cover slips in 24 well plates in maintenance media were switched to assay media (DMEM, 2% FBS, PSQ) (each subjects’ sample was run in duplicate). The next day, cells were treated with vehicle or quercetin (25μM) for 4 hr prior to the addition of cAMP+MPA or vehicle, as described above for decidualization assays. After 36 hr, cells were treated with GolgiPlug (BD Biosciences, San Jose, CA) to block IGFBP1 release and then 12 hr later (48 hr after vehicle or cAMP+MPA), cells were washed and fixed with 4% formaldehyde (freshly prepared), permeabilized/blocked with 5% mouse serum/0.3% Triton X-100/1X PBS, and stained with anti-IGFBP1-AF647 antibody (Santa Cruz, sc-55474 AF647, 1:100) and then stained with Alexa488-phalloidin (Abcam, 1:5000) and washed and mounted with Antifade reagent with DAPI (Fisher Scientific). Cells were then examined under a confocal microscope (Zeiss LSM900) using the 20x objective. ### Assessment of intracellular cAMP production by eSCs following forskolin or quercetin Intracellular cAMP ([cAMP]i) production by eSCs isolated from controls and endometriosis cases was analyzed using the cAMP ELISA kit (Cayman Chemical, Ann Arbor, MI, US) according to the manufacturer’s guidelines. Briefly, eSCs were grown to confluence and then seeded into 6-well plates at 3×105 cells/mL (2 mL/well) in maintenance media. 6 hr later, maintenance media was aspirated and replaced with 2% FBS-containing assay media. After an overnight incubation, eSCs were treated with IBMX (0.1mM, a phosphodiesterase inhibitor that blocks [cAMP]i degradation) for 30 min, followed by either vehicle, forskolin (25µM, an activator of adenylyl cyclases), or quercetin (25µM, as indicated) and incubated at 37°C with 5% CO2 using a method that was optimized in pilot studies. After 20 min, the media was aspirated and the cells were washed once with ice cold PBS and lysed with 0.25 mL 0.1N HCl for measuring [cAMP]i, as per the manufacturer’s directions. All samples were diluted 1:2 with ELISA diluent and run by ELISA assay in duplicate or triplicate. Data are expressed cAMP pMol/ml. ### Measurement of basal and H2O2-induced oxidative stress using eSCs Control- and endometriosis-eSCs isolated from controls were plated in black 96-well plates with clear bottoms (100 µL/well, 2.5X105/mL) in maintenance media. The numbers of subjects per group are shown in the figures. Media was aspirated and replaced with assay media and after an overnight incubation, cells were washed with HBSS, labeled with DCF-DA (20 µM) in HBSS for 30 min, and washed with HBSS 1% FBS (n=4-6 replicates per condition). Phenol red-free DMEM containing 2% FBS + PSQ (100µL/well) was then added, and cells were treated with either vehicle or quercetin (25µM) (or NAC, 10mM) and incubated at 37°C with 5% CO2 for 3 hr. Then cells were either treated with vehicle or H2O2 (500µM) and analyzed for oxidative stress 3 hr later in a quantitative manner using the VICTOR3 fluorescence plate reader (Perkin-Elmer) at an excitation wavelength of 485nm and emission wavelength of 535nm. This assay measures the conversion of non-fluorescent DCF-DA into fluorescent dichlorofluorescein (DCF) by reactive oxygen species (ROS) intermediates 75. Data are presented for each subject’s cells as percent vehicle-control oxidative stress according to condition (basal vs. induced). ### Blocking the effect of H2O2-inhibition of decidualization Testing the effect of quercetin on H2O2-impaired decidualization by eSCs was performed as described above, except eSCs in maintenance media were pre-treated with either vehicle or 250µM H2O2 for 2 hr and then washed. Maintenance media was replaced with 2% FBS-assay media. 48 hr later, eSCs were treated with vehicle or quercetin (25µM) for 4 hr and then stimulated with cAMP±MPA to induce decidualization and processed as above. ### Proteome profiler array Confluent control eSCs were plated in maintenance media in 100 mm plates and then switched to assay media 6 hr later. The next day, eSCs were treated with vehicle or quercetin (25µM) for 4 hr and then the cell lysates were processed and analyzed, as described by the Human Phospho-Kinase Array Panel Kit (ARY003C, R&D Systems). Image spots were quantified using the reference spots using ImageJ as described by the manufacturer. ### Western blotting Control-eSCs or endometriosis-eSCs were plated in maintenance media in 15 mm plates and, when confluent, the media was replaced with 2% FBS-assay media. The next day eSCs were treated with vehicle or quercetin (25 µM) for 4 hr. Cells were lysed in RIPA buffer containing protease and phosphatase inhibitors (Halt™ Protease and Phosphatase Inhibitor Cocktail, Thermo Fisher #PI78442) and incubated on ice for 25 min, followed by centrifugation at 13,000 rpm for 10 min at 4°C. Supernatants were collected and protein levels were quantified using the Pierce BCA Protein Assay Kit (Thermo Fisher, #23225); up to 65 µg/lane was run on 4-12% NuPAGE Bis-Tris 1.5 mm gels in NuPAGE MOPS running buffer (Invitrogen). Each lane represents an individual subject’s eSC lysate per condition. Proteins were transferred onto Immobilon FL-PVDF membranes, which were blocked at room temperature for 1 hr in PBS 0.1% Tween-20 (PBST) and 5% nonfat milk powder (#9999 CST). Membranes were rinsed in PBST and incubated overnight at 4°C in primary antibody (diluted 1:1000 in PBST containing 5% bovine serum albumin or 5% milk powder, depending on the manufacturer’s recommendation). See Supplementary Table S2 for listing of antibodies. The next day, membranes were washed in PBST and incubated for 1 hr at RT in HRP-conjugated secondary antibodies diluted in PBST containing 5% nonfat milk. Membranes were washed again in PBST and imaged by the addition of Clarity ECL substrate (#1705060, BioRad) or SuperSignal™ West Dura Extended Substrate (#34075, Thermo Fisher) on a ChemiDoc system (BioRad). SeeBlue™ Plus2 pre-stained molecular weight standard (#LC5925, Thermo Fisher) was used to estimate protein weight. Western blots were stripped with ReBlot Plus (#2504MI, Millipore), according to the manufacturer’s suggestions, and then re-blocked and probed as described above. ### Assessment of senescence markers by western blotting Briefly, eSCs were plated as described for western blotting above, except eSCs (p2-p3) in assay media were treated with vehicle or H2O2 (250 µM) for 2 hr. After the media was aspirated with replaced with assay media (DMEM 2% FBS, PSQ), eSCs were incubated for 2, 4, 6, or 8 days (as indicated) and then cell lysates were processed as described for western blotting using the Senescence Marker Antibody Sampler Kit (#56062, Cell Signaling Technology). Each lane represents an individual subject’s eSC lysates. For some experiments, eSCs were treated with vehicle or quercetin (Q, 25μM) for 4 hr post-H2O2 and analyzed for senescence markers at indicated times. ### scRNA-Seq analysis of fresh menstrual effluent Single-cell RNA sequencing (scRNA-Seq) data from whole, fresh ME, as previously described 25, was re-analyzed to determine *ADCY1-10* expression by ME-associated eSCs from control subjects. ### Assessment of apoptosis and senescence by flow cytometry #### Apoptosis Confluent cultures of control-eSCs (p3-p4) grown in maintenance media were switched to assay media and the following morning they were treated with vehicle or quercetin (25µM) for 24 or 48 hr (as indicated) prior to harvesting and processing for analysis of apoptosis by flow cytometry using FITC-Annexin V/PPI Apoptosis Detection Kit (#640914, BioLegend), according to the manufacturer’s directions. Flow cytometry data (fold-change after quercetin treatment) was acquired on a BD FACSymphony™ flow cytometer and analyzed using FlowJo™ v10 software. #### Senescence Confluent cultures of control-eSCs (p3-p4) were prepared as described above, except with the addition of 9H-(1,3-Dichloro-9,9-Dimethylacridin-2-One-7-yl) β-D-Galactopyranoside (DDAO galactoside or DDAOG) (#D6488,ThermoFisher), a substrate of SA-βgal that yields a far-red fluorescent product detected by flow cytometry at 670 nm, as previously described 76. Y-axis (FSC-A, size) and X-axis (DDAOG, 670nm). Sample data was acquired using the BD FACSymphony™ flow cytometer and analyzed using FlowJo™ v10 software. Viable cells were visualized in dot plots with DDAOG, 670nm versus FSC-A (size) and the percentage of large β-gal+ (senescent) cells was calculated. ### Statistics Analyses and graphical presentation were performed using the GraphPad Prism 9.0 or 10.0 software. Sample sizes for experiments are shown in each figure as individual data points. The numbers of assay replicates per subject or per condition (technical replicates) are indicated in each methods sections. The results are presented as the mean data for each subject’s cells (shown as individual data points); group data are shown as the mean ± SD (for parametric analyses [for normally distributed data with homogeneity of variance]) or the median ± interquartile range (IQR) (for non-parametric analyses [not normally distributed data without unequal variance]). Multiple comparisons were made by ANOVA with appropriate post-tests. Paired two group/sample data were analyzed using Wilcoxon matched-pairs signed rank test and unpaired two group/sample data (parametric) was analyzed by Mann Whitney U test. P values of 0.05 or less were considered significant (*p < 0.05; **p < 0.01; \***|p < 0.001; \**\*|\*p<0.0001). ## Data Availability All data produced in the present study are contained in the manuscript [https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE203191](https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE203191) ## Author contributions Julia G Delenko, Conceptualization, Experimentation/Methodology, Data curation, Validation, Formal analysis, Visualization, Writing – original draft, Writing – review and editing; Xiangying Xue, Experimentation/methodology, Data curation; Prodyot K Chatterjee, Experimentation/Methodology, Data curation, Validation, Formal Analysis, Visualization, Writing – original draft, Writing – review and editing; Nathaniel Hyman, Experimentation/Methodology, Data curation; Andrew J Shih, Formal analysis, Visualization, Writing – review and editing; Robert Adelson, Formal analysis, Visualization, Writing – review and editing; Polona Safaric-Tepes, Conceptualization, Experimentation/Methodology, Data curation, Validation, Writing – review and editing; Peter K Gregersen, Conceptualization, Formal analysis, Visualization, Resources, Writing – original draft, Writing – review and editing, Funding acquisition, Supervision; Christine N Metz, Conceptualization, Formal analysis, Visualization, Resources, Writing – original draft, Writing – review and editing, Funding acquisition, Supervision. All authors read and approved the final manuscript. ## Ethics statement Human subjects: The ROSE study for collecting menstrual effluent and menstrual effluent-derived endometrial stromal cells was reviewed and approved by the Institutional Review Board (IRB) of Northwell Health (IRB #13-376A). Written informed consent was obtained from all participants before sample collections and all samples were de-identified prior to processing. ## Competing Interests The authors declare that they have no competing interests. ## Funding This work was supported by the Northwell Health Innovations Award and the Endometriosis Foundation of America ## Data availability All data generated and analyzed during this study are included in the manuscript and are available. scRNA-Seq data describing *ADCY* transcript expression was obtained from AJ Shih (2022) deposited in the NCBI Gene Expression Omnibus, accession number GSE203191 ([https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE203191](https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE203191)). ## Additional files ### Supplementary files View this table: [Supplementary Table S1.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/T1) Supplementary Table S1. Primers for RT-qPCR View this table: [Supplementary Table S2.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/T2) Supplementary Table S2. Antibodies used for western blotting. ### Supplementary Figures ![](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F11/graphic-14.medium.gif) [](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F11/graphic-14) ![](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F11/graphic-15.medium.gif) [](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F11/graphic-15) Supplementary Figure S1. Quercetin enhances decidualization by control ME-eSCs as determined by immunofluorescence. **(A-B)** Control-eSCs (Control #1 (A) Control #2 (B)) were treated with vehicle or quercetin for 4 hr followed by vehicle or cAMP+MPA prior to immunofluorescent staining and confocal imaging. Merged images show IGFBP1 (red), phalloidin (green), and DAPI (blue) staining. IGFBP1 (at 20x magnification). IGFBP1+ cells are indicated by yellow arrows in the merged images. The scale bars (20 µm) are indicated. Single channel images are shown in black and white for IGFBP1, DAPI, and Phalloidin. ![Supplementary Figure S2.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F12.medium.gif) [Supplementary Figure S2.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F12) Supplementary Figure S2. Quercetin enhances decidualization by control-eSCs and endometriosis-eSCs, as determined by *IGFBP1* and *PRL* mRNA production. **(A-H)** Endometrial stromal cells (eSCs) from controls (A, C, E, and G, CTRL) and endometriosis patients (B, D, F, and H, ENDO) were treated with vehicle (Veh) or quercetin (Q, 25μM) for 4 hr prior to the addition of cAMP alone (A-B, E-F) or cAMP+MPA (C-D, G-H). After 48 hr, decidualization was analyzed by measuring *IGFBP1* (A-D) or *PRL* (E-H) mRNA expression by RT-qPCR (shown as fold-change in expression). Data points connected by a line represent paired data points from one individual’s eSCs (±Q). *p<0.05 Veh vs. Q-treated; **p<0.01 Veh vs. Q-treated. ![Supplementary Figure S3.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F13.medium.gif) [Supplementary Figure S3.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F13) Supplementary Figure S3. Quercetin enhances PGE2±MPA-stimulated decidualization. **(A-B)** Endometrial stromal cells (eSCs) from controls were treated with vehicle (Veh) or quercetin (Q, 25μM) for 4 hr prior to the addition of PGE2 alone (A) or PGE2+MPA (B). Decidualization was analyzed 48 hr later by measuring IGFBP1 protein by ELISA. Data points connected by a line represent paired data points from one individual’s eSCs (±Q). *p<0.05 Veh vs. Q-treated. ![Supplementary Figure S4.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F14.medium.gif) [Supplementary Figure S4.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F14) Supplementary Figure S4. Fresh menstrual effluent (ME) tissue-derived eSCs express multiple *ADCY* transcripts, as shown by scRNA-Seq. *ADCY* gene expression is shown from five different endometrial stromal cell (eSC) clusters (*IL11+, SRGN+, IGFBP1+, HSPA6+,* and *MGP+)* found in menstrual effluent (ME) 25. ![Supplementary Figure S5.](http://medrxiv.org/http://medrxiv.stage.highwire.org/content/medrxiv/early/2023/09/02/2023.08.30.23294800/F15.medium.gif) [Supplementary Figure S5.](http://medrxiv.org/content/early/2023/09/02/2023.08.30.23294800/F15) Supplementary Figure S5. Quercetin inhibits AKT signaling and promotes p53 (Ser46) phosphorylation in eSCs. Control-eSCs were treated with vehicle (Veh) or quercetin (25μM) for 4 hr before analysis using the Phospho-Kinase Array Panel (see upper right panel for key), as described in Figure 6 and in the methods section. ## Acknowledgements The authors would like to thank all of the participants of the ROSE study who donated menstrual effluent for the study, as well as the coordinators of the ROSE study (Margaret DeFranco, Kristine Sadicario, and Amber LaGuerre) for their help in recruiting, consenting, and enrolling research participants. In addition, we would like to thank Michael Ryan and the team in the Biorepository for maintaining and tracking cryopreserved endometrial stromal cells, as well as Laurianna Frasson and Rixsi Herrera for processing ME and isolating/freezing endometrial stromal cells and Amanda Chan, PhD, who assisted with the microscopy studies. We would like to thank JGD’s thesis committee members: Drs. Kim Simpfendorfer, Philippe Marambaud, and Myoungsun Son for their research insights and advice over the past two years. * Received August 30, 2023. * Revision received August 30, 2023. * Accepted September 2, 2023. * © 2023, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Gellersen, B., and Brosens, J.J. (2014). Cyclic decidualization of the human endometrium in reproductive health and failure. EndocrRev 35, 851–905. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1210/er.2014-1045&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25141152&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000346699200003&link_type=ISI) 2. 2.Barragan, F., Irwin, J.C., Balayan, S., Erikson, D.W., Chen, J.C., Houshdaran, S., Piltonen, T.T., Spitzer, T.L., George, A., Rabban, J.T., et al. (2016). Human Endometrial Fibroblasts Derived from Mesenchymal Progenitors Inherit Progesterone Resistance and Acquire an Inflammatory Phenotype in the Endometrial Niche in Endometriosis. Biol Reprod 94, 118. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1095/biolreprod.115.136010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27075616&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 3. 3.Klemmt, P.A., Carver, J.G., Kennedy, S.H., Koninckx, P.R., and Mardon, H.J. (2006). Stromal cells from endometriotic lesions and endometrium from women with endometriosis have reduced decidualization capacity. FertilSteril 85, 564–572. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.fertnstert.2005.08.046&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16500320&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000235978600004&link_type=ISI) 4. 4.Warren, L.A., Shih, A., Renteira, S.M., Seckin, T., Blau, B., Simpfendorfer, K., Lee, A., Metz, C.N., and Gregersen, P.K. (2018). Analysis of menstrual effluent: diagnostic potential for endometriosis. Mol Med 24, 1. 5. 5.Nayyar, A., Saleem, M.I., Yilmaz, M., DeFranco, M., Klein, G., Elmaliki, K.M., Kowalsky, E., Chatterjee, P.K., Xue, X., Viswanathan, R., et al. (2020). Menstrual Effluent Provides a Novel Diagnostic Window on the Pathogenesis of Endometriosis. Front Reprod Health 2, 3. 6. 6.Deryabin, P.I., and Borodkina, A.V. (2022). Stromal cell senescence contributes to impaired endometrial decidualization and defective interaction with trophoblast cells. Hum Reprod 37, 1505–1524. 7. 7.Brighton, P.J., Maruyama, Y., Fishwick, K., Vrljicak, P., Tewary, S., Fujihara, R., Muter, J., Lucas, E.S., Yamada, T., Woods, L., et al. (2017). Clearance of senescent decidual cells by uterine natural killer cells in cycling human endometrium. Elife 6. 8. 8.Khosla, S., Farr, J.N., Tchkonia, T., and Kirkland, J.L. (2020). The role of cellular senescence in ageing and endocrine disease. Nat Rev Endocrinol 16, 263–275. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41574-020-0335-y&link_type=DOI) 9. 9.Wilkinson, H.N., and Hardman, M.J. (2020). Senescence in Wound Repair: Emerging Strategies to Target Chronic Healing Wounds. Front Cell Dev Biol 8, 773. 10. 10.Paramos-de-Carvalho, D., Jacinto, A., and Saude, L. (2021). The right time for senescence. Elife 10. 11. 11.Kirkland, J.L., and Tchkonia, T. (2020). Senolytic drugs: from discovery to translation. J Intern Med 288, 518–536. 12. 12.Li, Y., Yao, J., Han, C., Yang, J., Chaudhry, M.T., Wang, S., Liu, H., and Yin, Y. (2016). Quercetin, Inflammation and Immunity. Nutrients 8, 167. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/nu8030167&link_type=DOI) 13. 13.Anand David, A.V., Arulmoli, R., and Parasuraman, S. (2016). Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn Rev 10, 84–89. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4103/0973-7847.194044&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28082789&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 14. 14. S.K. Shebeko, I.A.Z., O.S. Popov, O.O. Tarasenko, A.S. Shalamay. (2018). Effects of Quercetin and Its Combinations on Health.(London: Academic Press). 15. 15.Beazley, K.E., and Nurminskaya, M. (2016). Effects of dietary quercetin on female fertility in mice: implication of transglutaminase 2. Reprod Fertil Dev 28, 974–981. 16. 16.Vaez, S., Parivr, K., Amidi, F., Rudbari, N.H., Moini, A., and Amini, N. (2023). Quercetin and polycystic ovary syndrome; inflammation, hormonal parameters and pregnancy outcome: A randomized clinical trial. Am J Reprod Immunol 89, e13644. 17. 17.Jamali, N., Zal, F., Mostafavi-Pour, Z., Samare-Najaf, M., Poordast, T., and Dehghanian, A. (2021). Ameliorative Effects of Quercetin and Metformin and Their Combination Against Experimental Endometriosis in Rats. Reprod Sci 28, 683–692. 18. 18.Park, S., Lim, W., Bazer, F.W., Whang, K.Y., and Song, G. (2019). Quercetin inhibits proliferation of endometriosis regulating cyclin D1 and its target microRNAs in vitro and in vivo. J Nutr Biochem 63, 87–100. 19. 19.Zondervan, K.T., Becker, C.M., and Missmer, S.A. (2020). Endometriosis. N Engl J Med 382, 1244–1256. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMra1810764&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 20. 20.Halme, J., Hammond, M.G., Hulka, J.F., Raj, S.G., and Talbert, L.M. (1984). Retrograde menstruation in healthy women and in patients with endometriosis. Obstet Gynecol 64, 151–154. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=6234483&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1984TC63100001&link_type=ISI) 21. 21.Brosens, I., Brosens, J.J., and Benagiano, G. (2012). The eutopic endometrium in endometriosis: are the changes of clinical significance? Reprod Biomed Online 24, 496–502. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.rbmo.2012.01.022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22417665&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000303815500005&link_type=ISI) 22. 22.Bulun, S.E., Yilmaz, B.D., Sison, C., Miyazaki, K., Bernardi, L., Liu, S., Kohlmeier, A., Yin, P., Milad, M., and Wei, J. (2019). Endometriosis. Endocr Rev 40, 1048–1079. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1210/er.2018-00242&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30994890&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 23. 23.Vallve-Juanico, J., Houshdaran, S., and Giudice, L.C. (2019). The endometrial immune environment of women with endometriosis. Hum Reprod Update 25, 564–591. 24. 24.Liu, H., and Lang, J.H. (2011). Is abnormal eutopic endometrium the cause of endometriosis? The role of eutopic endometrium in pathogenesis of endometriosis. Med Sci Monit 17, RA92–99. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21455119&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 25. 25.Shih, A.J., Adelson, R.P., Vashistha, H., Khalili, H., Nayyar, A., Puran, R., Herrera, R., Chatterjee, P.K., Lee, A.T., Truskinovsky, A.M., et al. (2022). Single-cell analysis of menstrual endometrial tissues defines phenotypes associated with endometriosis. BMC Med 20, 315. 26. 26.Kalaitzopoulos, D.R., Samartzis, N., Kolovos, G.N., Mareti, E., Samartzis, E.P., Eberhard, M., Dinas, K., and Daniilidis, A. (2021). Treatment of endometriosis: a review with comparison of 8 guidelines. BMC Womens Health 21, 397. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12905-021-01545-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34844587&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 27. 27.Srivastava, S., Somasagara, R.R., Hegde, M., Nishana, M., Tadi, S.K., Srivastava, M., Choudhary, B., and Raghavan, S.C. (2016). Quercetin, a Natural Flavonoid Interacts with DNA, Arrests Cell Cycle and Causes Tumor Regression by Activating Mitochondrial Pathway of Apoptosis. Sci Rep 6, 24049. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/srep24049&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27068577&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 28. 28.Duo, J., Ying, G.G., Wang, G.W., and Zhang, L. (2012). Quercetin inhibits human breast cancer cell proliferation and induces apoptosis via Bcl-2 and Bax regulation. Mol Med Rep 5, 1453–1456. 29. 29.Kim, Y.J., Bae, Y.C., Suh, K.T., and Jung, J.S. (2006). Quercetin, a flavonoid, inhibits proliferation and increases osteogenic differentiation in human adipose stromal cells. Biochem Pharmacol 72, 1268–1278. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16996034&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 30. 30.Shi, J.W., Lai, Z.Z., Yang, H.L., Zhou, W.J., Zhao, X.Y., Xie, F., Liu, S.P., Chen, W.D., Zhang, T., Ye, J.F., et al. (2022). An IGF1-expressing endometrial stromal cell population is associated with human decidualization. BMC Biol 20, 276. 31. 31.Stadtmauer, D.J., and Wagner, G.P. (2022). Single-cell analysis of prostaglandin E2-induced human decidual cell in vitro differentiation: a minimal ancestral deciduogenic signaldagger. Biol Reprod 106, 155–172. 32. 32.Chen, M.M., Yin, Z.Q., Zhang, L.Y., and Liao, H. (2015). Quercetin promotes neurite growth through enhancing intracellular cAMP level and GAP-43 expression. Chin J Nat Med 13, 667–672. 33. 33.Burova, E., Borodkina, A., Shatrova, A., and Nikolsky, N. (2013). Sublethal oxidative stress induces the premature senescence of human mesenchymal stem cells derived from endometrium. Oxid Med Cell Longev 2013, 474931. 34. 34.Ticconi, C., Di Simone, N., Campagnolo, L., and Fazleabas, A. (2021). Clinical consequences of defective decidualization. Tissue Cell 72, 101586. 35. 35.Cinar, O., Seval, Y., Uz, Y.H., Cakmak, H., Ulukus, M., Kayisli, U.A., and Arici, A. (2009). Differential regulation of Akt phosphorylation in endometriosis. Reprod Biomed Online 19, 864–871. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.rbmo.2009.10.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20031030&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000273671000017&link_type=ISI) 36. 36.Madanes, D., Bilotas, M.A., Baston, J.I., Singla, J.J., Meresman, G.F., Baranao, R.I., and Ricci, A.G. (2020). PI3K/AKT pathway is altered in the endometriosis patient’s endometrium and presents differences according to severity stage. Gynecol Endocrinol 36, 436–440. 37. 37.Kim, T.H., Yu, Y., Luo, L., Lydon, J.P., Jeong, J.W., and Kim, J.J. (2014). Activated AKT pathway promotes establishment of endometriosis. Endocrinology 155, 1921–1930. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1210/en.2013-1951&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24605828&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 38. 38.Lee, Y.K., and Park, O.J. (2010). Regulation of mutual inhibitory activities between AMPK and Akt with quercetin in MCF-7 breast cancer cells. Oncol Rep 24, 1493–1497. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21042744&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 39. 39.Yoshino, O., Osuga, Y., Hirota, Y., Koga, K., Yano, T., Tsutsumi, O., and Taketani, Y. (2003). Akt as a possible intracellular mediator for decidualization in human endometrial stromal cells. Mol Hum Reprod 9, 265–269. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/molehr/gag035&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12728019&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000183575600004&link_type=ISI) 40. 40.Fabi, F., Grenier, K., Parent, S., Adam, P., Tardif, L., Leblanc, V., and Asselin, E. (2017). Regulation of the PI3K/Akt pathway during decidualization of endometrial stromal cells. PLoS One 12, e0177387. 41. 41.Gallolu Kankanamalage, S., Karra, A.S., and Cobb, M.H. (2018). WNK pathways in cancer signaling networks. Cell Commun Signal 16, 72. 42. 42.Lv, D., Guo, L., Zhang, T., and Huang, L. (2017). PRAS40 signaling in tumor. Oncotarget 8, 69076–69085. 43. 43.Havel, J.J., Li, Z., Cheng, D., Peng, J., and Fu, H. (2015). Nuclear PRAS40 couples the Akt/mTORC1 signaling axis to the RPL11-HDM2-p53 nucleolar stress response pathway. Oncogene 34, 1487–1498. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/onc.2014.91&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24704832&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 44. 44.Kim, W., Youn, H., Seong, K.M., Yang, H.J., Yun, Y.J., Kwon, T., Kim, Y.H., Lee, J.Y., Jin, Y.W., and Youn, B. (2011). PIM1-activated PRAS40 regulates radioresistance in non-small cell lung cancer cells through interplay with FOXO3a, 14-3-3 and protein phosphatases. Radiat Res 176, 539–552. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1667/RR2609.1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21910584&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 45. 45.Kusama, K., Yamauchi, N., Yoshida, K., Azumi, M., Yoshie, M., and Tamura, K. (2021). Senolytic treatment modulates decidualization in human endometrial stromal cells. Biochem Biophys Res Commun 571, 174–180. 46. 46.Voskarides, K., and Giannopoulou, N. (2023). The Role of TP53 in Adaptation and Evolution. Cells 12. 47. 47.Kang, H.J., and Rosenwaks, Z. (2018). p53 and reproduction. Fertil Steril 109, 39–43. 48. 48.Hu, W., Zheng, T., and Wang, J. (2011). Regulation of Fertility by the p53 Family Members. Genes Cancer 2, 420–430. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/1947601911408892&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21779510&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 49. 49.Paskulin, D.D., Cunha-Filho, J.S., Souza, C.A., Bortolini, M.C., Hainaut, P., and Ashton-Prolla, P. (2012). TP53 PIN3 and PEX4 polymorphisms and infertility associated with endometriosis or with post-in vitro fertilization implantation failure. Cell Death Dis 3, e392. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/cddis.2012.116&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23013791&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 50. 50.Embree-Ku, M., and Boekelheide, K. (2002). Absence of p53 and FasL has sexually dimorphic effects on both development and reproduction. Exp Biol Med (Maywood) 227, 545–553. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12094020&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 51. 51.Ochoa-Bernal, M.A., and Fazleabas, A.T. (2020). Physiologic Events of Embryo Implantation and Decidualization in Human and Non-Human Primates. Int J Mol Sci 21. 52. 52.Emera, D., Romero, R., and Wagner, G. (2012). The evolution of menstruation: a new model for genetic assimilation: explaining molecular origins of maternal responses to fetal invasiveness. Bioessays 34, 26–35. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/bies.201100099&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22057551&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 53. 53.Bonafe, M., Salvioli, S., Barbi, C., Trapassi, C., Tocco, F., Storci, G., Invidia, L., Vannini, I., Rossi, M., Marzi, E., et al. (2004). The different apoptotic potential of the p53 codon 72 alleles increases with age and modulates in vivo ischaemia-induced cell death. Cell Death Differ 11, 962–973. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.cdd.4401415&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15131588&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000223351300002&link_type=ISI) 54. 54.Dumont, P., Leu, J.I., Della Pietra, A.C., 3rd., George, D.L., and Murphy, M. (2003). The codon 72 polymorphic variants of p53 have markedly different apoptotic potential. Nat Genet 33, 357–365. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng1093&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12567188&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000181312700012&link_type=ISI) 55. 55.Kang, H.J., Feng, Z., Sun, Y., Atwal, G., Murphy, M.E., Rebbeck, T.R., Rosenwaks, Z., Levine, A.J., and Hu, W. (2009). Single-nucleotide polymorphisms in the p53 pathway regulate fertility in humans. Proc Natl Acad Sci U S A 106, 9761–9766. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTA2LzI0Lzk3NjEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wOS8wMi8yMDIzLjA4LjMwLjIzMjk0ODAwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 56. 56.Mohammadzadeh, M., Ghorbian, S., and Nouri, M. (2019). Evaluation of clinical utility of P53 gene variations in repeated implantation failure. Mol Biol Rep 46, 2885–2891. 57. 57.Palomares, A.R., Castillo-Dominguez, A.A., Ruiz-Galdon, M., Rodriguez-Wallberg, K.A., and Reyes-Engel, A. (2021). Genetic variants in the p53 pathway influence implantation and pregnancy maintenance in IVF treatments using donor oocytes. J Assist Reprod Genet 38, 3267–3275. 58. 58.Lledo, B., Turienzo, A., Ortiz, J.A., Morales, R., Ten, J., Llacer, J., and Bernabeu, R. (2014). Negative effect of P72 polymorphism on p53 gene in IVF outcome in patients with repeated implantation failure and pregnancy loss. J Assist Reprod Genet 31, 169–172. 59. 59.Liebl, M.C., and Hofmann, T.G. (2019). Cell Fate Regulation upon DNA Damage: p53 Serine 46 Kinases Pave the Cell Death Road. Bioessays 41, e1900127. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/bies.201900127&link_type=DOI) 60. 60.Deryabin, P., Griukova, A., Nikolsky, N., and Borodkina, A. (2020). The link between endometrial stromal cell senescence and decidualization in female fertility: the art of balance. Cell Mol Life Sci 77, 1357–1370. 61. 61.Hu, L., Li, H., Zi, M., Li, W., Liu, J., Yang, Y., Zhou, D., Kong, Q.P., Zhang, Y., and He, Y. (2022). Why Senescent Cells Are Resistant to Apoptosis: An Insight for Senolytic Development. Front Cell Dev Biol 10, 822816. 62. 62.Huang, W., Hickson, L.J., Eirin, A., Kirkland, J.L., and Lerman, L.O. (2022). Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol 18, 611–627. 63. 63.Kempuraj, D., Madhappan, B., Christodoulou, S., Boucher, W., Cao, J., Papadopoulou, N., Cetrulo, C.L., and Theoharides, T.C. (2005). Flavonols inhibit proinflammatory mediator release, intracellular calcium ion levels and protein kinase C theta phosphorylation in human mast cells. Br J Pharmacol 145, 934–944. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.bjp.0706246&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15912140&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000231252100011&link_type=ISI) 64. 64.Chirumbolo, S. (2010). The role of quercetin, flavonols and flavones in modulating inflammatory cell function. Inflamm Allergy Drug Targets 9, 263–285. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2174/187152810793358741&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20887269&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 65. 65.Kim, S.G., Sung, J.Y., Kim, J.R., and Choi, H.C. (2020). Quercetin-induced apoptosis ameliorates vascular smooth muscle cell senescence through AMP-activated protein kinase signaling pathway. Korean J Physiol Pharmacol 24, 69–79. 66. 66.Hohmann, M.S., Habiel, D.M., Coelho, A.L., Verri, W.A., Jr., and Hogaboam, C.M. (2019). Quercetin Enhances Ligand-induced Apoptosis in Senescent Idiopathic Pulmonary Fibrosis Fibroblasts and Reduces Lung Fibrosis In Vivo. Am J Respir Cell Mol Biol 60, 28–40. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1165/rcmb.2017-0289OC&link_type=DOI) 67. 67.Ozsoy Gokbilen, S., Becer, E., and Vatansever, H.S. (2022). Senescence-mediated anticancer effects of quercetin. Nutr Res 104, 82–90. 68. 68.Zoico, E., Nori, N., Darra, E., Tebon, M., Rizzatti, V., Policastro, G., De Caro, A., Rossi, A.P., Fantin, F., and Zamboni, M. (2021). Senolytic effects of quercetin in an in vitro model of pre-adipocytes and adipocytes induced senescence. Sci Rep 11, 23237. 69. 69.Bulun, S.E., Yildiz, S., Adli, M., Chakravarti, D., Parker, J.B., Milad, M., Yang, L., Chaudhari, A., Tsai, S., Wei, J.J., et al. (2023). Endometriosis and adenomyosis: shared pathophysiology. Fertil Steril 119, 746–750. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.fertnstert.2023.03.006&link_type=DOI) 70. 70.Kunz, G., Beil, D., Huppert, P., Noe, M., Kissler, S., and Leyendecker, G. (2005). Adenomyosis in endometriosis--prevalence and impact on fertility. Evidence from magnetic resonance imaging. Hum Reprod 20, 2309–2316. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/humrep/dei021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15919780&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000230725100039&link_type=ISI) 71. 71.Chaib, S., Tchkonia, T., and Kirkland, J.L. (2022). Cellular senescence and senolytics: the path to the clinic. Nat Med 28, 1556–1568. 72. 72.Signorile, P.G., Viceconte, R., and Baldi, A. (2018). Novel dietary supplement association reduces symptoms in endometriosis patients. J Cell Physiol 233, 5920–5925. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jcp.26401&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29243819&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 73. 73.Faber, J., and Fonseca, L.M. (2014). How sample size influences research outcomes. Dental Press J Orthod 19, 27–29. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1590/2176-9451.19.4.027-029.ebo&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25279518&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 74. 74.Repetto, G., del Peso, A., and Zurita, J.L. (2008). Neutral red uptake assay for the estimation of cell viability/cytotoxicity. Nat Protoc 3, 1125–1131. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nprot.2008.75&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18600217&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) 75. 75.Eruslanov, E., and Kusmartsev, S. (2010). Identification of ROS using oxidized DCFDA and flow-cytometry. Methods in molecular biology (Clifton, NJ) 594, 57–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/978-1-60761-411-1_4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20072909&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F09%2F02%2F2023.08.30.23294800.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000274938200004&link_type=ISI) 76. 76.Flor, A., Pagacz, J., Thompson, D., and Kron, S. (2022). Far-red Fluorescent Senescence-associated beta-Galactosidase Probe for Identification and Enrichment of Senescent Tumor Cells by Flow Cytometry. J Vis Exp.