Small extracellular vesicles derived from embryonic stem cells restore ovarian function of premature ovarian failure through PI3K/AKT signaling pathway

Mengyu Liu, Yu Qiu, Zhuowei Xue, Ruoyu Wu, Jie Li, Xin Niu, Ji Yuan, Yang Wang, Qingkai Wu, Mengyu Liu, Yu Qiu, Zhuowei Xue, Ruoyu Wu, Jie Li, Xin Niu, Ji Yuan, Yang Wang, Qingkai Wu

Abstract

Background: Premature ovarian failure (POF) has a great impact on reproductive endocrine function in females, and it is an important cause of infertility. Previous studies have demonstrated that small extracellular vesicles (sEVs) derived from stem cells play an important role in tissue regeneration. This study aimed to investigate the therapeutic effect of sEVs derived from embryonic stem cells (ESCs-sEVs) on damaged ovaries and explore the underlying molecular mechanisms.

Methods: Mice POF models were established by injecting mice with cyclophosphamide and busulfan. Then, ESCs-sEVs were intravenously transplanted into POF mice. The plasma of mice was harvested at 1 and 2 weeks after treatment to analyze the levels of anti-Mullerian hormone (AMH), estradiol (E2), and follicle stimulating hormone (FSH) by ELISA. The morphology of ovaries and follicles was observed by H&E staining, and apoptosis of granulosa cells was detected by TUNEL. In vitro, EdU and CCK-8 tests were used to evaluate the proliferation of cultured granulosa cells stimulated by ESCs-sEVs. Western blotting was used to determine the expression of PI3K/AKT and apoptotic-related proteins.

Results: After transplantation of ESCs-sEVs, the levels of serum sex hormones recovered to normal levels. In addition, the number of follicles was significantly increased, and the number of apoptotic cells was decreased. The results in vitro revealed that ESCs-sEVs could significantly improve the proliferation rate of granulosa cells and increase the expression of phosphorylated PI3K and AKT. Meanwhile, the positive effect on proliferation and the negative effect on apoptosis observed in granulosa cells were obviously decreased when the PI3K/AKT signaling pathway was inhibited.

Conclusion: Our findings suggested that ESCs-sEVs could improve ovarian function by regulating the PI3K/AKT signaling pathway, which could provide a promising clinical therapy for POF.

Keywords: Ovarian function; PI3K/AKT signaling pathway; Premature ovarian failure (POF); Small extracellular vesicle derived from embryonic stem cells (ESCs-sEVs).

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Characterization of ESCs and ESCs-sEVs. a Immunofluorescence detected the pluripotency markers in ESCs, including Oct-4, SSEA-4, Nanog, and TRA-1-81. Scale bars = 50 μm. b The morphology of ESCs-sEVs by TEM. Scale bars = 200 μm. c ESCs-sEVs were positive for CD9, CD63, and TSG101 and negative for GM130, Actin, and Lamin A/C, as shown by Western-blotting analysis. d Particle size distribution of ESCs-sEVs was determined by Flow Nano Analyzer
Fig. 2
Fig. 2
ESCs-sEVs contributed to the estrous cycle, body weight, and hormone levels in mice. A Estrous cycle of mice: (a) proestrus, (b) estrus, (c) metestrus, and (d) diestrus. B The weight of mice with ESCs-sEVs gradually increased to normal levels, while the weight of the CTX + BUS group gradually decreased to stable levels. The dashed line indicates mice that received treatment for 14 days. C E2 was significantly increased compared to the CTX + BUS group. D FSH was significantly decreased compared to the CTX + BUS group. E AMH was significantly increased compared to the CTX + BUS group. Scale bars = 100 μm. (Data are presented as the mean ± SD, *P < 0.05, **P < 0.01, ***P < 0.001; ns, no significance)
Fig. 3
Fig. 3
ESCs-sEVs promoted the development of follicles. A H&E staining of ovaries. A (e–h) Magnifications (× 100) of the red squares in A (a–d) (× 200), respectively. A (j) Primordial follicle. A (k) Primary follicle. A (l) Secondary follicle. A (m) Mature follicle. A (n) Atretic follicle. B Summary of follicles in each group. C Apoptosis was measured by TUNEL staining. The cell nuclei were stained by DAPI (blue fluorescence), and the apoptotic cells were stained with Cy3 (red fluorescence). D The percentage of TUNEL-positive granulosa cells was measured in each group. Apoptotic cells were increased in the CTX + BUS group and decreased in the ESCs-sEVs group. Scale bar = 75 μm E Western blotting analysis detected the expression of FSHR and a protein related to apoptosis in ovarian tissue. Data are presented as the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 4
Fig. 4
ESCs-sEVs promoted the proliferation and inhibited the apoptosis of ovary granulosa cells. A Morphology and identification of mouse ovarian granulosa cells. A (a) Granulosa cells adhered to the wall after 24 h (× 100). A (b) Granulosa cells proliferated significantly after 72 h (× 100). A (c) The phenotype of granulosa cells is shown by H&E staining (× 200), bars = 50 μm. A (d–f) Identification of ovarian granulosa cells by FSHR immunofluorescence (× 200). Bars = 100 μm. B ESCs-sEVs promoted the proliferation of granulosa cells, as detected by EdU tests; the cell nuclei are stained blue, and the cells with proliferation activity are stained green. C The proliferation ratio of granulosa cells was analyzed using EdU tests. D The cell proliferation curve of granulosa cells was measured by CCK-8 from day 1 to day 5. E The expression of FSHR, a protein related to apoptosis and the PI3K/AKT signaling pathway, was measured in granulosa cells by Western blotting analysis. Data are presented as the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 5
Fig. 5
The effect of ESCs-sEVs on granulosa cells was inhibited by a PI3K/AKT inhibitor. a ESCs-sEVs promoted the proliferation of granulosa cells, while the PI3K/AKT pathway inhibitor LY294002 reversed this effect. b The proliferation ratio of granulosa cells with/without ESCs-sEVs or LY294002. c CCK-8 tests indicated that the proliferation activity of granulosa cells was obviously inhibited in the presence of LY294002 as the time passed, which was consistent with the EdU test. d ESCs-sEVs increased the level of AMH synthesized by granulosa cells, while it was suppressed by LY294002. e The AMH secretion curve is shown in different groups at 24 h, 48 h, and 72 h. f ESCs-sEVs increased the phosphorylation of PI3K and AKT, as detected by Western blotting, and they inhibited the downstream apoptosis pathway. The results are presented as the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001, ESCs-sEVs versus control group. ###P < 0.001, ESCs-sEVs versus ESCs-sEVs + LY294002 group

References

    1. Kokcu A. Premature ovarian failure from current perspective. Gynecol Endocrinol. 2010;26(8):555–562. doi: 10.3109/09513590.2010.488773.
    1. Bao R, Xu P, Wang Y, et al. Bone marrow derived mesenchymal stem cells transplantation rescues premature ovarian insufficiency induced by chemotherapy. Gynecol Endocrinol. 2018;34(4):320–326. doi: 10.1080/09513590.2017.1393661.
    1. Li J, Yu Q, Huang H, et al. Human chorionic plate-derived mesenchymal stem cells transplantation restores ovarian function in a chemotherapy-induced mouse model of premature ovarian failure. Stem Cell Res Ther. 2018;9(1):81. doi: 10.1186/s13287-018-0819-z.
    1. Sheikhansari G, Aghebati-Maleki L, Nouri M, et al. Current approaches for the treatment of premature ovarian failure with stem cell therapy. Biomed Pharmacother. 2018;102:254–262. doi: 10.1016/j.biopha.2018.03.056.
    1. Yabut O, Bernstein HS. The promise of human embryonic stem cells in aging-associated diseases. Aging (Albany NY) 2011;3(5):494–508. doi: 10.18632/aging.100328.
    1. Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164(6):1226–1232. doi: 10.1016/j.cell.2016.01.043.
    1. Thery C, Witwer KW, Aikawa E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750. doi: 10.1080/20013078.2018.1535750.
    1. Matei Andreea C., Antounians Lina, Zani Augusto. Extracellular Vesicles as a Potential Therapy for Neonatal Conditions: State of the Art and Challenges in Clinical Translation. Pharmaceutics. 2019;11(8):404. doi: 10.3390/pharmaceutics11080404.
    1. Chen TS, Arslan F, Yin Y, et al. Enabling a robust scalable manufacturing process for therapeutic exosomes through oncogenic immortalization of human ESC-derived MSCs. J Transl Med. 2011;9:47. doi: 10.1186/1479-5876-9-47.
    1. Adamiak M, Cheng G, Bobis-Wozowicz S, et al. Induced pluripotent stem cell (iPSC)-derived extracellular vesicles are safer and more effective for cardiac repair than iPSCs. Circ Res. 2018;122(2):296–309. doi: 10.1161/CIRCRESAHA.117.311769.
    1. Chen B, Sun Y, Zhang J, et al. Human embryonic stem cell-derived exosomes promote pressure ulcer healing in aged mice by rejuvenating senescent endothelial cells. Stem Cell Res Ther. 2019;10(1):142. doi: 10.1186/s13287-019-1253-6.
    1. Wang Y, Yu D, Liu Z, et al. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res Ther. 2017;8(1):189. doi: 10.1186/s13287-017-0632-0.
    1. Zhang Q, Xu M, Yao X, et al. Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility. Stem Cell Res Ther. 2015;6:152. doi: 10.1186/s13287-015-0148-4.
    1. Adhikari D, Risal S, Liu K, et al. Pharmacological inhibition of mTORC1 prevents over-activation of the primordial follicle pool in response to elevated PI3K signaling. PLoS One. 2013;8(1):e53810. doi: 10.1371/journal.pone.0053810.
    1. Santos JMS, Lins T, Barberino RS, et al. Kaempferol promotes primordial follicle activation through the phosphatidylinositol 3-kinase/protein kinase B signaling pathway and reduces DNA fragmentation of sheep preantral follicles cultured in vitro. Mol Reprod Dev. 2019;86(3):319–329. doi: 10.1002/mrd.23107.
    1. Maclaran K, Panay N. Premature ovarian failure. J Fam Plann Reprod Health Care. 2011;37(1):35–42. doi: 10.1136/jfprhc.2010.0015.
    1. Jankowska K. Premature ovarian failure. Prz Menopauzalny. 2017;16(2):51–56.
    1. Sarosiek KA, Ni Chonghaile T, Letai A. Mitochondria: gatekeepers of response to chemotherapy. Trends Cell Biol. 2013;23(12):612–619. doi: 10.1016/j.tcb.2013.08.003.
    1. Kalich-Philosoph L, Roness H, Carmely A, et al. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med. 2013;5(185):185ra162. doi: 10.1126/scitranslmed.3005402.
    1. Shirai Makoto, Sakurai Ken, Saitoh Wataru, Matsuyama Takuya, Teranishi Munehiro, Furukawa Tadashi, Sanbuissho Atsushi, Manabe Sunao. Collaborative work on evaluation of ovarian toxicity 8) Two- or four-week repeated-dose studies and fertility study of Anastrozole in female rats. The Journal of Toxicological Sciences. 2009;34(Special):SP91–SP99. doi: 10.2131/jts.34.S91.
    1. Lai D, Wang F, Yao X, et al. Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure. J Transl Med. 2015;13:155. doi: 10.1186/s12967-015-0516-y.
    1. Fu X, He Y, Xie C, et al. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy. 2008;10(4):353–363. doi: 10.1080/14653240802035926.
    1. Yu YS, Sui HS, Han ZB, et al. Apoptosis in granulosa cells during follicular atresia: relationship with steroids and insulin-like growth factors. Cell Res. 2004;14(4):341–346. doi: 10.1038/sj.cr.7290234.
    1. Wang W, Luo SM, Ma JY, et al. Cytotoxicity and DNA damage caused from diazinon exposure by inhibiting the PI3K-AKT pathway in porcine ovarian granulosa cells. J Agric Food Chem. 2019;67(1):19–31. doi: 10.1021/acs.jafc.8b05194.
    1. Sun Y, Lin Y, Li H, et al. 2,5-Hexanedione induces human ovarian granulosa cell apoptosis through BCL-2, BAX, and CASPASE-3 signaling pathways. Arch Toxicol. 2012;86(2):205–215. doi: 10.1007/s00204-011-0745-7.
    1. Sasson R, Amsterdam A. Stimulation of apoptosis in human granulosa cells from in vitro fertilization patients and its prevention by dexamethasone: involvement of cell contact and bcl-2 expression. J Clin Endocrinol Metab. 2002;87(7):3441–3451. doi: 10.1210/jcem.87.7.8676.
    1. Lakhani SA, Masud A, Kuida K, et al. Caspases 3 and 7: key mediators of mitochondrial events of apoptosis. Science. 2006;311(5762):847–851. doi: 10.1126/science.1115035.
    1. Yin N, Wang Y, Lu X, et al. hPMSC transplantation restoring ovarian function in premature ovarian failure mice is associated with change of Th17/Tc17 and Th17/Treg cell ratios through the PI3K/Akt signal pathway. Stem Cell Res Ther. 2018;9(1):37. doi: 10.1186/s13287-018-0772-x.
    1. Tsuruta F, Masuyama N, Gotoh Y. The phosphatidylinositol 3-kinase (PI3K)-Akt pathway suppresses Bax translocation to mitochondria. J Biol Chem. 2002;277(16):14040–14047. doi: 10.1074/jbc.M108975200.

Source: PubMed

3
購読する