Abstract Senescence of bone marrow mesenchymal stem cells (BMMSCs) induced by chronic oxidative stress is an important factor contributes to the postmenopausal osteoporosis (PMOP). Mitochondrial quality control takes a pivotal role in regulating oxidative stress and cell senescence. Genistein is a major isoflavone in soy products, which is best known for its ability to inhibit bone loss in both postmenopausal women and ovariectomized (OVX) rodents. Here we show that OVX-BMMSCs displayed premature senescence, elevated reactive oxygen species (ROS) level and mitochondria dysfunction, while genistein rescued these phenotypes. Using network pharmacology and molecular docking, we identified estrogen-related receptor α (ERRα) as the potential target of genistein. Knockdown of ERRα greatly abolished the anti-senescence effect of genistein on OVX-BMMSCs. Further, the mitochondrial biogenesis and mitophagy induced by genistein were inhibited by ERRα knockdown in OVX-BMMSCs. In vivo, genistein inhibited trabecular bone loss and p16^INK4a expression, upregulated sirtuin 3 (SIRT3) and peroxisome proliferator-activated receptor gamma coactivator one alpha (PGC1α) expression in the trabecular bone area of proximal tibia in OVX rats. Together, this study revealed that genistein ameliorates senescence of OVX-BMMSCs through ERRα-mediated mitochondrial biogenesis and mitophagy, which provided a molecular basis for advancement and development of therapeutic strategies against PMOP. Keywords: Mesenchymal stem cells, Senescence, Mitophagy, Estrogen-related receptor α, Osteoporosis 1. Introduction Postmenopausal osteoporosis (PMOP) is a systemic skeletal disorder characterized by low bone mass and increased fracture risk, which is triggered by estrogen deficiency during menopause [[43]1]. Premature senescence of stem cells is recognized as a critical pathogenesis of bone deteriorations in PMOP [[44]2]. Mechanistically, mitochondrial dysfunctions, in particular bioenergetic decline and reactive oxygen species (ROS) accumulation, are crucial mechanisms underlying stem cell senescence. Accordingly, approaches aimed at restoring mitochondrial homeostasis may provide promising opportunities to counteract PMOP [[45]3]. Mitochondrial homeostasis is a sophisticated regulatory network that coordinates various cellular pathways, including the interplay between mitochondrial biogenesis and mitophagy. The tight coordination of these two opposing processes preserves mitochondrial function and cell homeostasis. Mitochondrial biogenesis provides the cell with newly synthesized mitochondria, thus preserves mitochondrial function and cellular homeostasis. Inhibition of mitochondrial biogenesis can lead to defective osteogenesis of bone marrow mesenchymal stem cells (BMMSCs) [[46]4]. Besides mitochondrial biogenesis, degradation of defective or superfluous mitochondria through mitophagy process also serves as another machinery for the maintenance of mitochondrial integrity and quality control [[47]5]. Defective mitophagy can lead to accumulation of damaged mitochondria [[48]6], which generate inefficient oxidative phosphorylation and further increases ROS production. Induction of mitophagy alleviates the damage caused by oxidative stress [[49]7]. Therefore, restoring the mitochondrial homeostasis of BMMSCs is possibly an effective method to against PMOP. Genistein, the principal soy phytoestrogen, has been proved to prevent bone loss in postmenopausal women [[50]8] and ovariectomized (OVX) rodents [[51]9,[52]10]. It was reported that genistein could mitigate mitochondrial dysfunctions in many pathological conditions, including neuroinflammation and aggregation of Aβ in Alzheimer's disease [[53]11]. Therefore, it is likely that genistein exert anabolic effect partially through modulating mitochondrial homeostasis to reduce oxidative stress, thus reverse premature senescence of OVX-BMMSCs during estrogen deficiency. In the present study, we systematically characterized the role of genistein in regulating the senescence of BMMSCs during estrogen deficiency. We determined that OVX-BMMSCs presented premature senescence phenotype compared to the Sham-BMMSCs, and genistein treatment greatly ameliorated cellular senescence in OVX-BMMSCs. Using integrated bioinformatic analysis, we identified estrogen-related receptor α (ERRα) as the potential target of genistein in PMOP. Remarkably, genistein-induced mitochondria biogenesis and mitophagy were largely prevented by transfection of ERRα siRNA. As a result, the anti-senescence effect of genistein in OVX-BMMSCs was abolished by ERRα knockdown. Collectively, these data demonstrate genistein exert anti-senescence effect on OVX-BMMSCs by restoring mitochondrial homeostasis in an ERRα-dependent manner, which provide further evidence for its use as a natural therapeutic agent for PMOP. It also suggested that the agonists of ERRα may be a viable therapeutic strategy to preserve bone mass in PMOP. 2. Materials and methods 2.1. Cell culture and reagents Twelve-week-old female Sprague-Dawley rats were subjected to either bilateral ovariectomy or sham surgery. Briefly, rats were anaesthetized and the bilateral ovaries were gently removed via a dorsal approach. For the Sham group, animals received the operation without removing the ovaries. Sixteen weeks after surgery, Sham-BMMSCs and OVX-BMMSCs were harvested from the bone marrow of the tibias and femurs from Sham rats and OVX rats, respectively. Sham-BMMSCs were maintained in DMEM medium plus 10% FBS (Gibco, Grand Island, NY, USA) and 1% penicillin-streptomycin (Gibco). OVX-BMMSCs were cultured in the same condition except that the DMEM medium is phenol red free (Gibco) to mimic the environment in vivo. Genistein and N-acetylcysteine (NAC) were obtained from Sigma (Sigma-Aldrich Inc., St. Louis, MO, USA) and bafilomycin A1 (Baf-A1) was purchased from CST (Cell Signaling Technology, Beverly, MA, USA). NAC is a classic antioxidant that is well known for its ability to minimize intracellular oxidative stress. Baf-A1, a vacuolar-type H^+-translocating ATPase (V-ATPase) inhibitor, was used as an autophagy inhibitor as it blocks the fusion of autophagosomes with lysosomes [[54]12]. Antibodies were from various sources, including Abcam (anti-p21^CIP1, anti-TOM20, anti-PGC1α, anti-LC3B, anti-P62, HRP-conjugated goat anti-rabbit and FITC-conjugated goat anti-rabbit), CST (anti-p53, anti-CoxIV, anti-γH2AX, anti-β-actin, HRP-conjugated anti-rabbit and HRP-conjugated anti-mouse), Invitrogen (anti-p16^INK4a, anti-SIRT3 and anti-LepR), and Proteintech (anti-PGC1α and anti-Parkin). 2.2. Phenotypic characterization For phenotypic characterization, cells were stained with fluorochrome-conjugated monoclonal antibodies including CD45-FITC (Elabscience, E-AB-F1227C), CD31-FITC (Abcam, ab33858), CD90-FITC (Elabscience, E-AB-F1226C) and CD44-FITC (Elabscience, E-AB-F1225C). Corresponding mouse IgG1-FITC (Elabscience, E-AB-F09792C) and mouse IgG2a-FITC (Elabscience, E-AB-F09802C) were used as isotype control. The samples were incubated for 30 min in the dark and then washed for 2 times. All samples were resuspended in 500 μl PBS and analyzed by flow cytometer (Beckman, CytoFLEX LX) and FlowJo software (Version 10.8.1, Tree Star, San Carlos, CA). 2.3. Small interfering RNA (siRNA) transfection ERRα siRNAs and negative control (NC) siRNAs were purchased from HanBio Technology Company (Shanghai, China) and the transfection was carried out according to the manufacturer's instruction. Briefly, cells were seeded in 6-well plate using growth medium without antibiotic one day before transfection. ERRα siRNA or NC siRNA were premixed with Lipofectamine RNAiMAX (Invitrogen) and then added into the cell medium with the final siRNA concentration of 100 nM. After 48-h incubation, the transfection medium was changed with fresh medium with or without genistein. The protein expression of ERRα in cells transfected with siRNA was examined by western blot analysis. The sequences of rat siRNA are as follows: ERRα: 5ʹ- GGCCUCCAAUGAGUGUGAGAU-3ʹ; Scrambled siRNA: 5ʹ-UUCUCCGAACGUGUCACGU-3ʹ. 2.4. Cell viability assay Cell viability was evaluated by cell counting kit-8 (Dojindo, Japan). Briefly, 3 × 10^3 cells were plated in 96 well plates and were incubated with various concentrations of genistein for 24 h, 48 h and 72 h. After treatment, cells were incubated in 10% CCK-8 reagent for another 2 h. The optical density (OD) value was measured at 450 nm with a microplate reader (ELx800; BioTek Instruments, USA). 2.5. Colony-forming assays Cells (300 cells/well) were seeded into 6-well plates and cultured in conditioned medium for 10 days. Cells were fixed in 4% paraformaldehyde (m/v) for 15min at room temperature (RT). After rinsing with PBS, cells were stained with crystal violet solution (Sigma). The images were captured by microscope and the colonies (aggregates≥50 cells) were counted. 2.6. Senescence β-galactosidase (β-Gal) staining To assess cell senescence, β-Gal activity was determined by using β-Gal Staining Kit (Beyotime, Institute of Biotechnology, China). Briefly, cells were fixed in fixation buffer for 15 min at RT, then washed and incubated with staining solution at 37 °C overnight. The number of blue cells positive for SA-β-Gal was expressed as % of total cell number (100 cells counted for each condition, three independent experiments). 2.7. Immunofluorescence staining Cells were seeded onto gelatin-coated glass slides and treated with conditioned medium for 48 h. After removing the medium, the cells were fixed with 4% paraformaldehyde for 20 min, permeabilized with 0.2% Triton X-100 for 10min, and then blocked with goat serum for 1 h. For protein detection and localization, the cells were incubated with specific primary antibodies (anti-γH2AX, 1:400; anti-Parkin, 1:200; anti-TOM20, 1:200) overnight. All samples were then washed 3 times and incubated with fluorescent secondary antibody (1:1000, Abcam) for 2 h. DAPI was used for nucleus counter staining. The photographs were acquired by using a Nikon fluorescence microscope. The γH2AX intensity was divided by the DAPI intensity. Three random fields of view were included into the measurement for each slide, and the assays were repeated for three biological replicates. 2.8. Mitochondrial DNA copy number assay Mitochondrial DNA (mtDNA) copy number was determined by quantitative real-time PCR as described [[55]13]. Briefly, total DNA was extracted and purified from cells using a DNeasy Blood & Tissue kit (Qiagen, Hilden, Germany). Quantification of mtDNA was performed by use of the ratio of mtDNA marker mitochondrial NADH dehydrogenase 1 (mt-ND1) (Forward: 5′-CGTTGCCCAAACCATCTCT-3′, Reverse: 5′-CTATTGGTCAGGCGGGGA-3′) to nuclear DNA marker β-actin (Forward: 5′- TTGTCACCAACTGGGACGATATGG-3′, Reverse: 5′- GGGGTGTTGAAGGTCTCAAACATG-3′). The relative mtDNA copy number was measured by normalization of the crossing points in quantitative PCR curves between ND1 and β-actin genes using the RelQuant software (Roche Applied Sciences). 2.9. ROS measurements Cellular ROS level was measured by a DCFDA fluorescent probe (Beyotime). Briefly, cells were wash with HBSS twice and incubated with serum-free DMEM containing 10 μM DCFDA at 37 °C for 20 min. The photographs were captured by a Nikon fluorescence microscope. ROS generation was determined by using a FACSCalibur flow cytometer (Beckman, CytoFLEX LX). The data from flow cytometry were analyzed using FlowJo software, and mean fluorescence intensity (MFI) in each group was recorded and analyzed. When indicated, 5 mM NAC was used as an antioxidant positive control. 2.10. Mitochondrial superoxide Quantification Cells were incubated with 5 μM MitoSOX Red (Invitrogen) for 10 min, then washed with warm HBSS to remove the excess dye. To visualize the stained cells, random fields in each group were captured by using a Nikon fluorescence microscope. To quantify the MitoSOX Red fluorescence intensity, cells were collected and sorted by FACSCalibur flow cytometer (Beckman). The data from flow cytometry were analyzed using FlowJo software, and the mean fluorescence intensity (MFI) in each group was recorded and analyzed. 2.11. ATP content and mitochondrial membrane potential detection ATP content of cells was determined with the ATP assay kit (Abcam, ab83355) according to manufacturer's protocol. Briefly,1 × 10^6 cells were resuspended in 100 μL of ATP assay buffer, then homogenized and centrifuged for 5 min at 4 °C. The supernatant was collected and added to plate at 50 μL/well. Add 50 μL ATP reaction mix to each well, incubate the plate at room temperature for 30 min, then measure output on a microplate reader at OD 570 nm. Subtract the mean value of the blank from all standards and sample readings. The sample values were calculated from the standard curve and normalized to the control group in each individual experiment. For measuring mitochondrial membrane potential, a JC-1 kit (Beyotime) was used. JC-1 is cationic dye that exhibit potential-dependent accumulation in mitochondria, indicated by a fluorescence emission shift from green to red. Mitochondrial depolarization is indicated by a decrease in the red/green fluorescence intensity ratio. Cells were incubated with JC-1 dye for 20 min at 37 °C in the dark. Thereafter, cells were either assessed by fluorescence microscope or harvested and analyzed by flow cytometry. The data from flow cytometry were analyzed using FlowJo software, and median PE and FITC fluorescence were used to calculate a red/green ratio as an index of mitochondrial membrane potential [[56]14]. 2.12. Oxygen consumption rate (OCR) The measurement of OCR in cells were performed using Seahorse XF96 analyzer (Agilent Technologies, Santa Clara, CA) following manufacturer's protocol. Briefly, OVX-BMMSCs were plated at density of 10,000 cells/well in a Seahorse cell culture microplate and transfected with siNC/siERRα. After 48 h transfection, the media is replaced with culturing medium with/without genistein for another 72 h. On the day of measurement, the medium was changed to pre-warmed Seahorse assay medium, and OCR determined using the Seahorse XF Cell Mito Stress Kit (Agilent). After recording 3 total cellular respiration measurements in unstimulated cells, oligomycin (1.5 μM) was added to inhibit mitochondrial ATP synthase and measure the decrease in the OCR that is linked to ATP turnover. To determine the maximal respiration potential of the cells, FCCP (1 μM; an uncoupler of oxidative phosphorilation) was used. The amount of nonmitochondrial oxygen consumption was determined by inhibiting the respiratory chain activity with antimycin A and rotenone cocktail (0.5 μM). After completion, cells were lysed with RIPA buffer (Beyotime), and OCR was normalized to protein content determined using a BCA protein assay kit (Beyotime) according to the manufacturer's instructions. 2.13. Transmission electron microscopy (TEM) analysis Cells were washed with PBS (PH 7.4) and primarily fixed in 2.5% glutaraldehyde for 24 h. The samples were post-fixed in 1% OsO[4] for 1 h at RT, then progressively dehydrated through graded ethanol/acetone solutions and embedded in epoxy resin. Finally, ultrathin sections (70 nm) were prepared and stained with 2% uranyl acetate followed by lead citrate. The images of cell ultrastructure were captured with a transmission electron microscope (JEOL, Tokyo, Japan). The experiments were repeated for three biological replicates. 2.14. Live cell imaging microscopy Cells were loaded with green-fluorescing mitochondrial dye Mito-Tracker Green (MTG, 20 nM) (Invitrogen) for 30min at 37 °C, then washed 2 times with fresh complete growth medium and incubated with red-fluorescing LysoTracker Red (LTR, 50 nM) (Invitrogen) for 30min at 37 °C. After MTG and LTR had been loaded, confocal images were acquired by using a confocal laser scanning microscope (TCS SP8, Leica Microsystems, Germany). MTG was excitated at 490/516 nm and LTR was excitated at 577/590 nm. Co-localization of both signals is counted as a mitophagy event. The Pearson's colocalization coefficient between MTG and LTR fluorescence intensities was determined in 9 cells from three independent experiments using Image J software [[57]13]. For functional mitochondria staining, cells were loaded with Mito-Tracker Red CMXRos (MTR, 20 nM) (Invitrogen) for 30min at 37 °C, then washed 2 times with fresh complete growth medium. Confocal images were acquired by using a confocal laser scanning microscope (Leica). MTR was excitated at 579/599 nm. The stained cells were also sorted by FACSCalibur flow cytometer (Beckman). The data from flow cytometry were analyzed using FlowJo software, and the mean fluorescence intensity (MFI) in each group was recorded and analyzed. 2.15. Total RNA extraction and quantitative real-time PCR analysis Targeted gene expression analyses were performed by rt-qPCR as described [[58]15]. Briefly, total RNA was extracted with Trizol reagent (Invitrogen, Carlsbad, CA), followed by cDNA synthesis using Reverse Transcription Kit (TaKaRa, Japan) according to the manufacturer's instructions. qRT-PCR were performed on a Roche Light Cycler 480 (Roche, Mannheim, Germany) using the SYBR Green PCR Master Mix (TaKaRa, Japan). Fold changes of mRNA were calculated by the 2^−ΔΔCt method after normalization to the expression of β-actin (ACTB). Sequences of the primers are listed in [59]Supplementary Table S1. 2.16. Western blotting The cells were dissociated with RIPA buffer (Beyotime) containing protease inhibitor and phosphatase inhibitors (Sigma). After concentration measurement, 20 μg of protein was loaded onto 4–20% SDS-PAGE gel and transferred to a PVDF membrane (Millipore, Billerica, MA, USA). The membrane was blocked by 5% non-fat milk for 2 h followed by incubated with primary antibody at 4 °C overnight. Subsequently, the blots were incubated for 1 h at RT with an HRP-conjugated secondary antibody (CST), and visualized by chemiluminescence detection system (Millipore). Quantitative analysis of the Western blot was carried out by using Image J software, and band intensity was normalized to β-actin. The primary antibodies used were, anti-p21^CIP1 (1:1000, Abcam, ab109199) anti-TOM20 (1:1000, Abcam, ab186735), anti-PGC1α (1 μg/ml, Abcam, ab106814), anti-LC3B (1:2000, Abcam, ab192890), anti-P62 (1:1000, Abcam, ab56416), anti-p53 (1:1000, CST, #2524), anti-CoxIV (1:1000, CST, #4850), anti-β-actin (1:1000, CST, #8457), and anti-SIRT3 (1:1000, Invitrogen, PA5-96406). 2.17. Drug-target interaction prediction Drug–target network (DTN) from genistein and targets associations in postmenopausal osteoporosis was constructed and analyzed by the DrugBank 4.0 database. Then, we mapped the primary target proteins into the online STRING database to generate protein-protein interaction (PPI) network generation. Interaction pairs with overall score above 0.9 were recorded. To characterize the biochemical pathways and functions linked to genistein, GO (Gene Ontology) enrichment analysis and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis were carried out. 2.18. Molecular docking analysis The chemical structure of genistein was obtained from the PubChem database, and genistein were imported into Schrodinger for 3D format conversion and energy minimization. The protein structure of ERRα was obtained by SwisS-Model online server and processed in Maestro11.9. Schrodinger's Protein Preparation Wizard was used to process the protein structure, remove the crystal water, replenish the missing hydrogen atoms, repair the missing bond information, repair the missing peptide, and finally minimize the energy of the Protein [[60]16,[61]17]. Then, Molecular docking studies using ERRα with genistein was performed by the Glide module in Schrodinger Maestro software. Finally, the screening and analysis of docking results were carried out by SP. 2.19. Animals and drug treatment All animal protocol was critically reviewed and approved by the institutional review board of Shanghai Jiao Tong University School of Medicine (institutional review board no. SH9H-2020-A704-1). This study conformed to the ARRIVE (Animal Research: Reporting In Vivo Experiments) guidelines for animal studies. A total of 60 female Sprague-Dawley rats, mean weight 220 ± 25 g, were obtained at 3-month of age from the Ninth People's Hospital Animal Center (Shanghai, China). Rats were maintained under the following environmental conditions: temperature: 25 ± 3 °C; humidity: 50 ± 10%; illumination: light/dark cycle 12/12; free access to water and a diet low in phytoestrogen content. Following 2 weeks of adaptation, 30 rats were received operation to establish the OVX-OP model and 30 rats were received sham operation. Eight weeks after surgery, rats received the following treatments with 15 rats in each group: (1) Sham + VEH group: oral saline solution treatment once daily; (2) Sham + GEN group: oral genistein (50 mg/kg body weight, Sigma-Aldrich, MO, USA) treatment once daily. (3) OVX + VEH group: oral saline solution treatment once daily; (4) OVX + GEN group: oral genistein (50 mg/kg body weight) treatment once daily. After being fed for 8 weeks, blood was withdrawn before the rats were sacrificed, and serum was then prepared. Rats were sacrificed by cervical dislocation under halothane anesthesia. Femurs and tibias were collected for further analysis. 2.20. Measurement of serum MDA The activity of MDA in serum was measured by using a lipid peroxidation MDA assay kit (Beyotime) according to the manufacturer's instructions. Briefly, the serum was incubated with reagent in each reaction tube at 100 °C for 15 min. After centrifuged at 4000 rpm for 10 min, the supernatants were collected and measured for absorbance at 532 nm with a microplate reader. 2.21. Bone biomechanical test The femurs were stored frozen at −20 °C before biomechanical test. After thawing, the right femur mid-shaft was taken for the three-point bending assay by using electronic universal testing machine (Shenzhen Reger Instrument Co. Ltd., China) according to Ma et al.'s [[62]18] description. A load was vertically applied to the femoral mid-shaft, with a displacement speed of 0.1 mm/s and span distance at 20 mm until the femoral shaft is fractured. The left femoral neck was tested by mounting the proximal half of the femur vertically in a chuck and applying a downward force on the femoral head until failure. The ultimate load of the femurs was recorded and analyzed. 2.22. Micro-computed tomography (Micro-CT) Micro-CT scanning was performed by using a micro-CT system (μCT-80, Scanco Medical, Bassersdorf, Switzerland) on proximal tibias before decalcification. To visualize the inside of the proximal tibia, two-dimensional (2-D) images at their most central part were obtained. Trabecular bone was selected as region of interests (ROI) and the following parameters of ROI were analyzed: bone mineral density (BMD), bone volume/total volume (BV/TV), trabecular thickness (Tb.Th) and trabecular separation (Tb.Sp). 2.23. Histological analysis Decalcification of specimens was carried out with 10% EDTA with constant shaking for 8 weeks. After decalcification, the specimens were embedded in paraffin and cut into 5-μm-thick sections. Three sections of the most central part of the proximal tibia were selected and underwent H-E staining. For immunohistochemistry, sections were deparaffinized, rehydrated, and incubated with primary antibody (anti-p16^INK4a, 5 μg/mL, Invitrogen) at 4 °C overnight after antigen retrieval. The sections were then washed in PBS and incubated with secondary antibody (HRP-conjugated goat anti-rabbit, 1:1000, Abcam), and stained with DAB kit. The sections were observed under a light microscope. For immunofluorescence staining, sections were incubated with a primary antibody (anti-SIRT3, 1:100, Invitrogen) at 4 °C overnight and then with a secondary antibody (FITC-conjugated goat anti-rabbit, 1:1000, Abcam) for 1 h at 37 °C. Afterwards, slides were stained with DAPI and mounted with an anti-fade reagent (Invitrogen). Confocal images of slides were obtained by using a confocal microscope (Leica). For peroxisome proliferator-activated receptor gamma coactivator one alpha (PGC1α) and leptin receptor (LepR) immunofluorescence staining, sections were blocked with 10% goat serum (Gibco) and then incubated with primary antibodies (anti- PGC1α, 1:50, Proteintech; anti-LepR, 1:50, Invitrogen) at 4 °C overnight and with secondary antibodies for 1 h at 37 °C. Slides were then stained with DAPI and mounted with an anti-fade reagent (Invitrogen). The images were captured by a Zeiss LSM710 confocal microscope (Carl Zeiss). Image J software was used for quantification of IHC and IF. For quantification of p16^INK4a staining, three random fields of view per specimen were taken, and the average values were taken as the data point for each specimen. The SIRT3 intensity was divided by the DAPI intensity, and the PGC1α intensity was divided by the LepR intensity. Three random fields of view were included into the measurement for each specimen, and nine specimens in each group were selected for immunohistochemistry and immunofluorescence staining. All data was analyzed blinded. 2.24. Statistical analysis Data were presented as means ± SD. At least three samples or independent experiments were performed with all the assays. All statistical analyses were performed using the GraphPad prism (Version 9.0, GraphPad Software, San Diego, CA). Normal distribution of the data was verified using Shapiro-Wilk test and the equality of variances was verified by Levene's test. P values were calculated by unpaired Student's t-test, or one-way ANOVA for samples following normal distribution. For ANOVA, Tukey's post-test was used to compare individual groups. Mann-Whitney, or Kruskal-Wallis tests were used when samples didn't follow a normal distribution. For Kruskal-Wallis test, Dunn's multiple comparisons test was used for post-hoc analysis. P value < 0.05 was taken as statistical significance. 3. Results 3.1. Genistein alleviated the senescence of OVX-BMMSCs Primary cultured BMMSCs were positive for surface markers (CD44 and CD90) and negative for hematopoietic cell markers (CD31 and CD45) ([63]Fig. 1A). Cell viability assay showed that genistein had no cell toxicity when used at concentrations ranging from 10^−3 to 1 μM ([64]Fig. 1B). The expression of phosphorylated H2A histone family member X (γH2AX), a marker of DNA damage, was upregulated in the OVX-BMMSCs versus Sham-BMMSCs ([65]Fig. 1C and D). Consistently, OVX-BMMSCs displayed senescence phenotype, as shown by the reduced colony forming ability ([66]Fig. 1E and F) and increased SA-β-gal positive cells ([67]Fig. 1E, G). Then, we explored the effect of genistein on senescence of OVX-BMMSCs at concentration of 1 μM and 10^−2μM, as these concentrations have been identified as the optimal concentrations in previous study [[68]19]. Administration of genistein could increase the number and size of colonies, and downregulate the SA-β-gal and γH2AX positive cells ([69]Fig. 1C–G). We also analyzed the mRNA and protein expressions of senescence related marker [[70]20] p53, p16^INK4a and p21^CIP1, and found that the OVX-induced upregulation of p53, p16^INK4a and p21^CIP1 in BMMSCs were inhibited by genistein treatment ([71]Fig. 1H and I). Collectively, these data indicated that the estrogen-deficiency induced premature senescence could be alleviated by genistein treatment in BMMSCs. Fig. 1. [72]Fig. 1 [73]Open in a new tab Genistein alleviated cellular senescence of OVX-BMMSCs. (A) Flow cytometric analysis for cell surface markers of rat BMMSCs (n = 3). (B) Cell viability assay of cells treated by escalating doses of genistein (0–10 μM) for 24 h, 48 h, and 72 h (n = 3). (C, D) Representative images of γH2AX staining and quantitative assessment of the γH2AX positive cells (n = 3). DAPI-labeled nuclei are in blue and γH2AX is stained in green. Cells were treated by genistein for 3 days. Scale bar, 50 μm. (E, F and G) The representative images and quantitative analysis of colony forming assay and SA-β-gal staining (n = 3). Scale bar, 500 μm. (H, I) The mRNA and protein expression of senescence related marker p53, p16^INK4a and p21^CIP1 of Sham-BMMSCs and OVX-BMMSCs. Cells were treated by genistein for 3 days, when indicated. Statistical analysis of expression of each marker was adjusted to β-actin (n = 3). Each value is mean ± SD. Significance was calculated using a one-way ANOVA followed by the Tukey's post-hoc test (B, D, G, H and I). Kruskal-Wallis test followed by Dunn's multiple comparisons test was used for F. *p < 0.05 and **p < 0.01 compared to the Sham group. #p < 0.05 and ##p < 0.01 compared to the OVX group. (For interpretation of the references to color in this figure legend, the