Exosomes derived from human adipose mesenchymal stem cells improve ovary function of premature ovarian insufficiency by targeting SMAD

Boxian Huang, Jiafeng Lu, Chenyue Ding, Qinyan Zou, Wei Wang, Hong Li, Boxian Huang, Jiafeng Lu, Chenyue Ding, Qinyan Zou, Wei Wang, Hong Li

Abstract

Background: Although many reports show that various kinds of stem cells have the ability to recover the function of premature ovarian insufficiency (POI), few studies are associated with the mechanism of stem cell treatment of POI. We designed this experimental study to investigate whether human adipose stem cell-derived exosomes (hADSC-Exos) retain the ability to restore ovarian function and how hADSC-Exos work in this process.

Methods: A POI mouse model was established and human ovarian granule cells (hGCs) collected from individuals with POI were prepared to assess the therapeutic effects and illuminate the mechanism of hADSCs in curing POI. The hematoxylin and eosin assay method was employed to assess the number of follicles. Enzyme-linked immunosorbent assay (ELISA) was used to detect the serum levels of sex hormones. The proliferation rate and marker expression levels of hGCs were measured by flow cytometry (fluorescence-activated cell sorting). Real-time PCR and western blot assays were used to determine the mRNA and protein expression levels of SMAD2, SMAD3, and SMAD5. Western blot assays were used to test the protein expression levels of apoptosis genes (Fas, FasL, caspase-3, and caspase-8).

Results: After the hADSC-Exos were transplanted into the POI mice model, they exerted better therapeutic activity on mouse ovarian function, improving follicle numbers during four stages. ELISA results showed that hADSC-Exos elevated the hormone levels to the normal levels. In addition, after hADSC-Exos were cocultured with POI hGCs, our results showed that hADSC-Exos significantly promoted the proliferation rate and inhibited the apoptosis rate. Furthermore, hADSC-Exos also increased the marker expression of hGCs to the normal level. Besides, mRNA and protein assays demonstrated that hADSC-Exos downregulated the expression of SMAD2, SMAD3, and SMAD5 in vivo and in vitro. Western blot assay demonstrated that hADSC-Exos inhibited expression of the apoptosis genes in POI hGCs, and SMAD knockdown increased the protein expression of apoptosis genes.

Conclusions: These findings demonstrate for the first time the molecular cascade and related cell biology events involved in the mechanism by which exosomes derived from hADSCs improved ovarian function of POI disease via regulation of the SMAD signaling pathway.

Keywords: Exosome; Human adipose stem cells; Premature ovarian insufficiency; SMAD pathway.

Conflict of interest statement

The use of human ovarian granular cells and human peripheral blood mononuclear cells was in accordance with the relevant guidelines and regulations, and the experimental protocols were approved by the Medical Ethics Committee of the Suzhou Hospital Affiliated to Nanjing Medical University (NJMU-2015-014). All of the patients provided written informed consent prior to participation in this study. Our investigation using experimental animals was conducted on the basis of the Nanjing Medical University Animal Center’s specific guidelines and standards.

Not applicable.

The authors declare that they have no competing interests.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Characterization of hADSC-Exos. a Phenotype of hADSC-Exos detected by electron microscopy. b Expression levels of CD9, CD63, and CD81 in hADSC-Exos detected by flow cytometry. c Expression levels of CD9, CD63, and CD81 in hADSC-Exos detected by western blot. Scale bars = 100 nm. GAPDH glyceraldehyde 3-phosphate dehydrogenase, hADSC human adipose mesenchymal stem cells
Fig. 2
Fig. 2
hADSC-Exos improved function of POI mouse model. a Number of primordial follicles counted over 4 weeks after hADSC-Exos injection. b Number of primary follicles counted over 4 weeks after hADSC-Exos injection. c Number of secondary follicles counted over 4 weeks after hADSC-Exos injection. d Number of antral follicles counted over 4 weeks after hADSC-Exos injection. All experiments carried three times; error bars indicate SD. **p < 0.01, ***p < 0.001 (compared with POI group). POI premature ovarian insufficiency, PBS phosphate buffered saline
Fig. 3
Fig. 3
hADSC-Exos improved hormone level of POI mouse model. a E2 levels measured by ELISA over 4 weeks after hADSC-Exos injection. b FSH levels measured by ELISA over 4 weeks after hADSC-Exos injection. c AMH levels measured by ELISA over 4 weeks after hADSC-Exos injection. All experiments carried three times; error bars indicate SD. ***p < 0.001 (compared with POI group). E2 estradiol, FSH follicle-stimulating hormone, AMH anti-Mullerian hormone, POI premature ovarian insufficiency
Fig. 4
Fig. 4
hADSC-Exos improve proliferation rate and inhibit apoptosis rate in POI hGCs. a Schematic overview of hGC filter procedures. b hADSC-Exos improve proliferation in hGCs more significantly than using PBS. c hADSC-Exos inhibit apoptosis in hGCs more effectively than using PBS. Error bars indicate SD. ***p < 0.001 (compared with normal group). GC granulosa cell, POI premature ovarian insufficiency, hGC human granulosa cell, PBS phosphate buffered saline
Fig. 5
Fig. 5
hADSC-Exos improve marker expression in POI hGCs. a hADSC-Exos increase number of FSHR+AMH+ hGCs. b hADSC-Exos increase number of FOXL2+CYP19A1+ hGCs. Error bars indicate SD. **p < 0.01, ***p < 0.001 (compared with normal group). POI premature ovarian insufficiency, PBS phosphate buffered saline, EXO exosome, FSHR follicle-stimulating hormone receptor, AMH anti-Mullerian hormone
Fig. 6
Fig. 6
hADSC-Exos upregulated SMAD pathway in POI hGCs. a qPCR analysis of mRNA expression levels of SMAD2, SMAD3, and SMAD5 after hADSC-Exos coculture with POI hGCs. b Western blot analysis of protein expression levels of SMAD2, SMAD3, and SMAD5 after hADSC-Exos coculture with POI hGCs. Error bars indicate SD. ***p < 0.001 (compared with normal group); ###p < 0.001 (compared with hADSC-Exos treatment group). POI premature ovarian insufficiency, PBS phosphate buffered saline, hADSC human adipose mesenchymal stem cells, GAPDH glyceraldehyde 3-phosphate dehydrogenase, EXO exosome
Fig. 7
Fig. 7
hADSC-Exos upregulated SMAD pathway in POI mouse model. a qPCR analysis of mRNA expression levels of SMAD2, SMAD3, and SMAD5 after hADSC-Exos injection into POI mouse model. b Western blot analysis of protein expression levels of SMAD2, SMAD3, and SMAD5 after injection into POI mouse model. Error bars indicate SD. ***p < 0.001 (compared with normal group); ###p < 0.001 (compared with hADSC-Exos treatment group). POI premature ovarian insufficiency, PBS phosphate buffered saline, hADSC human adipose mesenchymal stem cells, GAPDH glyceraldehyde 3-phosphate dehydrogenase, EXO exosome
Fig. 8
Fig. 8
hADSC-Exos repress apoptosis genes through SMAD pathway. a Western blot analysis of protein expression levels of Fas, FasL, Caspase-8, and Caspase-3 after hADSC-Exos coculture with hGCs. b Western blot analysis of protein expression levels of Fas, FasL, Caspase-8, and Caspase-3 after SMAD2, SMAD3, and SMAD5 knockdown in normal hGCs. c Proposed model for exosomes derived from hADSCs improved ovaries of POI mice through regulating SMAD pathway. Error bars indicate SD. **p < 0.01, ***p < 0.001 (compared with normal group). GAPDH glyceraldehyde 3-phosphate dehydrogenase, POI premature ovarian insufficiency, PBS phosphate buffered saline, EXO exosome, siRNA small interfering RNA, hADSC human adipose mesenchymal stem cells

References

    1. Ding C, Li H, Wang Y, Wang F, Wu H, Chen R, Lv J, Wang W, Huang B. Different therapeutic effects of cells derived from human amniotic membrane on premature ovarian aging depend on distinct cellular biological characteristics. Stem Cell Res Ther. 2017;8:173. doi: 10.1186/s13287-017-0613-3.
    1. Pascuali N, Scotti L, Di Pietro M, Oubina G, May M, Gomez Munoz A, Cuasnicu PS, Cohen DJ, Tesone M, Abramovich D, Parborell F. Ceramide-1-phosphate has protective properties against cyclophosphamide-induced ovarian damage in a mice model of premature ovarian failure. Hum Reprod 2018. Epub ahead of print.
    1. Preventive Services Task Force US, Grossman DC, Curry SJ, Owens DK, Barry MJ, Davidson KW, Doubeni CA, Epling JW, Jr KAR, Krist AH, Kurth AE, Landefeld CS, Mangione CM, Phipps MG, Silverstern M, Simon MA, Tseng CW. Hormone therapy for the primary prevention of chronic conditions in postmenopausal women: US Preventive Services Task Force recommendation statement. JAMA. 2017;318:2224–2233. doi: 10.1001/jama.2017.18261.
    1. Ding C, Zou Q, Wang F, Wu H, Wang W, Li H, Huang B. HGF and BFGF secretion by human adipose-derived stem cells improves ovarian function during natural aging via activation of the SIRT1/FOXO1 signaling pathway. Cell Physiol Biochem. 2018;45:1316–1332. doi: 10.1159/000487559.
    1. Ding C, Zou Q, Wang F, Wu H, Chen R, Lv J, Ling M, Sun J, Wang W, Li H, Huang B. Human amniotic mesenchymal stem cells improve ovarian function in natural aging through secreting hepatocyte growth factor and epidermal growth factor. Stem Cell Res Ther. 2018;9:55. doi: 10.1186/s13287-018-0781-9.
    1. Su J, Ding L, Cheng J, Yang J, Li X, Yan G, Sun Hm Dai J, Hu Y. Transplantation of adipose-derived stem cells combined with collagen scaffolds restores ovarian function in a rat model of premature ovarian insufficiency. Hum Reprod. 2016;31:1075–1086. doi: 10.1093/humrep/dew041.
    1. Takehara Y, Yabuuchi A, Ezoe K, Kuroda T, Yamadera R, Sano C, Murata N, Aida T, Nakama K, Aono F, Aoyama N, Kato K, Kato O. The restorative effects of adipose-derived mesenchymal stem cells on damaged ovarian function. Lab Investig. 2013;93:181–193. doi: 10.1038/labinvest.2012.167.
    1. Sun M, Wang S, Li Y, Yu L, Gu F, Wang C, Yao Y. Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure. Stem Cell Res Ther. 2013;4:80. doi: 10.1186/scrt231.
    1. Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol Ther. 2015;23:812–823. doi: 10.1038/mt.2015.44.
    1. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659. doi: 10.1038/ncb1596.
    1. Yang PC. Induced pluripotent stem cell (iPSC)-derived exosomes for precision medicine in heart failure. Circ Res. 2018;122:661–663. doi: 10.1161/CIRCRESAHA.118.312657.
    1. Chen L, Xiang B, Wang X, Xiang C. Exosomes derived from human menstrual blood-derived stem cells alleviate fulminant hepatic failure. Stem Cell Res Ther. 2017;8:9. doi: 10.1186/s13287-016-0453-6.
    1. Meyer K, Yankner S. Slowing down aging. Cell Metab. 2017;26:592–593. doi: 10.1016/j.cmet.2017.09.012.
    1. Li Q, Inhibitory SMAD. Potential regulators of ovarian function. Biol Reprod. 2015;92:50. doi: 10.1095/biolreprod.114.125203.
    1. Tripurani SK, Cook RW, Eldin KW, Pangas SA. BMP-specific SMADs function as novel repressors of PDGFA and modulate its expression in ovarian granulosa cells and tumors. Oncogene. 2013;32:3877–3885. doi: 10.1038/onc.2012.392.
    1. Coss D, Mellon PL, Thackray VG. A FoxL in the Smad house: activin regulation of FSH. Trends Endocrinol Metab. 2010;21:562–568. doi: 10.1016/j.tem.2010.05.006.
    1. Fortin J, Boehm U, Weinstein MB, Graff JM, Bernard DJ. Follicle-stimulating hormone synthesis and fertility are intact in mice lacking SMAD3 DNA binding activity and SMAD2 in gonadotrope cells. FASEB J. 2014;28:1474–1485. doi: 10.1096/fj.13-237818.
    1. Whyte J, Glover JD, Woodcock M, Brzeszczynska J, Taylor L, Sherman A, Kaiser P, McGrew MJ. FGF, insulin, and SMAD signaling cooperate for avian primordial germ cell self-renewal. Stem Cells Rep. 2015;5:1171–1182. doi: 10.1016/j.stemcr.2015.10.008.
    1. Nie M, Yu S, Peng S, Fang Y, Wang H, Yang X. miR-23a and miR-27a promote human granulosa cell apoptosis by targeting SMAD5. Biol Reprod. 2015;93:98. doi: 10.1095/biolreprod.115.130690.
    1. Ding C, Zou Q, Ding J, Ling M, Wang W, Li H, Huang B. Increased N6-methyladenosine causes infertility is associated with FTO expression. J Cell Physiol. 2018;233:7055–7066. doi: 10.1002/jcp.26507.
    1. Wang Y, Fortin J, Lamba P, Bonomi M, Persani L, Roberson MS, Bernard DJ. Activator protein-1 and Smad proteins synergistically regulate human follicle-stimulating hormone β-promoter activity. Endocinology. 2008;149:5577–5591. doi: 10.1210/en.2008-0220.
    1. Eke G, Mangir N, Hasirci N, MacNeil S, Hasirci V. Development of a UV crosslinked biodegradable hydrogel containing adipose derived stem cells to promote vascularization for skin wounds and tissue engineering. Biomaterials. 2017;129:188–198. doi: 10.1016/j.biomaterials.2017.03.021.
    1. Choi HS, Kim HJ, Oh JH, Park HG, Ra JG, Chang KA, Suh YH. Therapeutic potentials of human adipose-derived stem cells on the mouse model of Parkinson’s disease. Neurobiol Aging. 2015;36:2885–2892. doi: 10.1016/j.neurobiolaging.2015.06.022.
    1. Li F, Turan V, Lierman S, Cuvelier C, De Sutter P, Oktay K. Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death. Hum Reprod. 2014;29:107–113. doi: 10.1093/humrep/det391.
    1. Zhang Q, Bu S, Sun J, Xu M, Yao X, He K, Lai D. Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage. Stem Cell Res Ther. 2017;8:270. doi: 10.1186/s13287-017-0721-0.
    1. Hong S, Kim HY, Kim J, Ha HT, Kim YM, Bae E, Kim TH, Lee KC, Kim SJ. Smad7 protein induces interferon regulatory factor 1-dependent transcriptional activation of caspase 8 to restore tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis. J Biol Chem. 2013;288:3560–3570. doi: 10.1074/jbc.M112.400408.

Source: PubMed

3
订阅