Human amniotic mesenchymal stem cells improve the follicular microenvironment to recover ovarian function in premature ovarian failure mice

Rongxia Liu, Xiaoyu Zhang, Zhenhai Fan, Yuying Wang, Guanping Yao, Xue Wan, Zulin Liu, Bing Yang, Limei Yu, Rongxia Liu, Xiaoyu Zhang, Zhenhai Fan, Yuying Wang, Guanping Yao, Xue Wan, Zulin Liu, Bing Yang, Limei Yu

Abstract

Background: Many adult women younger than 40 years old have premature ovarian failure (POF) and infertility. Previous studies confirmed that different tissue-derived stem cells could restore ovarian function and folliculogenesis in chemotherapy-induced POF mice. The aim of this study was to explore the therapeutic efficacy and underlying mechanisms of human amniotic mesenchymal stem cells (hAMSCs) transplantation for hydrogen peroxide-induced ovarian damage.

Methods: Bilateral ovaries of female mice were burned with 10% hydrogen peroxide to establish a POF model. After 24 h of treatment, hAMSCs and diethylstilbestrol were administered to POF mice by intraperitoneal injection and intragastric administration, respectively. After either 7 or 14 days, ovarian function was evaluated by the oestrus cycle, hormone levels, ovarian index, fertility rate, and ovarian morphology. The karyotype was identified in offspring by the G-banding technique. hAMSCs tracking, immunohistochemical staining, and real-time polymerase chain reaction (PCR) were used to assess the molecular mechanisms of injury and repair.

Results: The oestrus cycle was recovered after hAMSCs transplantation at 7 and 14 days. Oestrogen levels increased, while follicle-stimulating hormone levels decreased. The ovarian index, fertility rate, and population of follicles at different stages were significantly increased. The newborn mice had no obvious deformity and showed normal growth and development. The normal offspring mice were also fertile. The tracking of hAMSCs revealed that they colonized in the ovarian stroma. Immunohistochemical and PCR analyses indicated that changes in proteins and genes might affect mature follicle formation.

Conclusions: These results suggested that hAMSCs transplantation can improve injured ovarian tissue structure and function in oxidatively damaged POF mice. Furthermore, the mechanisms of hAMSCs are related to promoting follicular development, granulosa cell proliferation, and secretion function by improving the local microenvironment of the ovary.

Keywords: Diethylstilbestrol; Human amniotic mesenchymal stem cells; Hydrogen peroxide; Microenvironment; Ovary; Premature ovarian failure; Reproduction.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Morphology and molecular phenotype of hAMSCs. The cells morphology of passage 0 (a, × 40), 3 and 4 (b, c, × 100) hAMSCs. d Phenotypic analysis of the fourth-passage hAMSCs by flow cytometry
Fig. 2
Fig. 2
The transplanted hAMSCs improve oestrus cycles in POF mice. Vaginal smears were obtained, and the oestrus cycles were evaluated by H&E staining. Representative photographs for proestrus (a), oestrus (b), metoestrus (c), and dioestrus (d) are shown (× 100). e The percentage of abnormal cyclicity was detected in mice at 7 and 14 days after cells transplantation (n = 6)
Fig. 3
Fig. 3
Transplantation of hAMSCs improves serum oestrogen and FSH hormone levels in POF mice. Serum levels of oestrogen (a) and FSH (b) were detected at 7 and 14 days after cells transplantation. Data are presented as the mean ± S (x¯ ± s, n = 6), *P < 0.05
Fig. 4
Fig. 4
The body weight and ovarian index of POF mice were obviously increased by transplanted hAMSCs. Body weight (a) and ovarian index (b) were detected at 7 and 14 days after cells transplantation (x¯ ± s, n = 6). *P < 0.05
Fig. 5
Fig. 5
Transplanted hAMSCs improved the fertility rate in POF mice. a The different fertility rates are shown in each group, but the DES group showed infertility (n = 10). b The suckling mice of the hAMSC group were observed for 30 days. Karyotype analysis of the chromosomes of offspring mice in the normal group (c) and hAMSC group (d) were observed under oil immersion lenses (× 400)
Fig. 6
Fig. 6
Transplantation of hAMSCs reduces ovarian injuries in POF mice. a The pathological changes of ovaries were evaluated by H&E staining in the four groups at 7 and 14 days after hAMSCs transplantation (× 40). Quantitation of follicle count from ovaries in mice of the four groups at 7 (b) and 14 (c) days after cell transplantation (n = 4). The black arrow shows granulosa cells, the red arrow shows the cumulus complex, and the green arrow shows mature follicles
Fig. 7
Fig. 7
In vivo hAMSCs tracking. a, b PKH26-labelled hAMSCs showed red fluorescence (× 100). c The labelling rates of PKH26-labelled hAMSCs were detected by flow cytometry. Transplanted hAMSCs were observed at 7 (d, e) and 14 (f, g) days after cell transplantation in ovaries (× 100, n = 3). d and f show the bright field under a fluorescence microscope, and e and g show the dark field (× 100)
Fig. 8
Fig. 8
The hAMSCs improved protein expression associated with follicular development in the ovarian microenvironment of POF mice. The brown particles of positive expression were observed on FSHR, VEGF, IGF-1, TNF-α, and IL-1β proteins by immunohistochemical staining at 7 days (a) and 14 days (c) after hAMSCs transplantation. At 7 (b) and 14 days (d) after cell transplantation, the mean integral optical density value (IOD) of FSHR-, VEGF-, IGF-1-, TNF-α-, and IL-1β-positive areas was determined in ovarian tissue by Image-Pro Plus (immunohistochemical staining, × 200). Data are presented as the mean ± S (x¯ ± s, n = 6), *P < 0.05
Fig. 9
Fig. 9
The mRNA levels of FOXL2, OCT4, GDF-9, SCF, and LIF were regulated by hAMSCs in the ovarian tissues of POF mice by real-time PCR at 7 (a) and 14 (b) days after cells transplantation. Data are presented as the mean ± S (x¯ ± s, n = 6), *P < 0.05

References

    1. Fenton AJ. Premature ovarian insufficiency: pathogenesis and management. J Midlife Health. 2015;6:147–153.
    1. Jankowska K. Premature ovarian failure. Prz Menopauzalny. 2017;16:51–56.
    1. Kovanci E, Schutt AK. Premature ovarian failure: clinical presentation and treatment. Obstet Gynecol Clin N Am. 2015;42:153–161. doi: 10.1016/j.ogc.2014.10.004.
    1. Sullivan SD, Sarrel PM, Nelson LM. Hormone replacement therapy in young women with primary ovarian insufficiency and early menopause. Fertil Steril. 2016;106:1588–1599. doi: 10.1016/j.fertnstert.2016.09.046.
    1. Yin N, Zhao W, Luo Q, Yuan W, Luan X, Zhang H. Restoring ovarian function with human placenta-derived mesenchymal stem cells in autoimmune-induced premature ovarian failure mice mediated by treg cells and associated cytokines. Reprod Sci. 2018;25:1073–1082. doi: 10.1177/1933719117732156.
    1. Park M, Suh DS, Lee K, Bae J. Positive cross talk between FOXL2 and antimüllerian hormone regulates ovarian reserve. Fertil Steril. 2014;102:847–855. doi: 10.1016/j.fertnstert.2014.05.031.
    1. de Castro FC, Cruz MH, Leal CL. Role of growth differentiation factor 9 and bone morphogenetic protein 15 in ovarian function and their importance in mammalian female fertility - a review. Asian-Australas J Anim Sci. 2016;29:1065–1074. doi: 10.5713/ajas.15.0797.
    1. Atala A. Engineering tissues, organs and cells. J Tissue Eng Regen Med. 2007;1:83–96. doi: 10.1002/term.18.
    1. Sun L, Li D, Song K, Wei J, Yao S, Li Z, et al. Exosomes derived from human umbilical cord mesenchymal stem cells protect against cisplatin-induced ovarian granulosa cell stress and apoptosis in vitro. Sci Rep. 2017;7:2552. doi: 10.1038/s41598-017-02786-x.
    1. Kilic S, Pinarli F, Ozogul C, Tasdemir N, Naz Sarac G, Delibasi T. Protection from cyclophosphamide-induced ovarian damage with bone marrow-derived mesenchymal stem cells during puberty. Gynecol Endocrinol. 2014;30:135–140. doi: 10.3109/09513590.2013.860127.
    1. Liu J, Zhang H, Zhang Y, Li N, Wen Y, Cao F, et al. Homing and restorative effects of bone marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats. Mol Cells. 2014;37:865–872. doi: 10.14348/molcells.2014.0145.
    1. Gabr H, Rateb MA, El Sissy MH, Ahmed Seddiek H, Ali Abdelhameed Gouda S. The effect of bone marrow-derived mesenchymal stem cells on chemotherapy induced ovarian failure in albino rats. Microsc Res Tech. 2016;79:938–947. doi: 10.1002/jemt.22725.
    1. Badawy A, Sobh MA, Ahdy M, Abdelhafez MS. Bone marrow mesenchymal stem cell repair of cyclophosphamide-induced ovarian insufficiency in a mouse model. Int J Women's Health. 2017;9:441–447. doi: 10.2147/IJWH.S134074.
    1. Fu X, He Y, Wang X, Peng D, Chen X, Li X, et al. Overexpression of miR-21 in stem cells improves ovarian structure and function in rats with chemotherapy-induced ovarian damage by targeting PDCD4 and PTEN to inhibit granulosa cell apoptosis. Stem Cell Res Ther. 2017;8:187. doi: 10.1186/s13287-017-0641-z.
    1. Liu T, Huang Y, Guo L, Cheng W, Zou G. CD44+/CD105+ human amniotic fluid mesenchymal stem cells survive and proliferate in the ovary long-term in a mouse model of chemotherapy-induced premature ovarian failure. Int J Med Sci. 2012;9:592–602. doi: 10.7150/ijms.4841.
    1. Lai D, Wang F, Chen Y, Wang L, Wang Y, Cheng W. Human amniotic fluid stem cells have a potential to recover ovarian function in mice with chemotherapy-induced sterility. BMC Dev Biol. 2013;13:34. doi: 10.1186/1471-213X-13-34.
    1. Xiao GY, Liu IH, Cheng CC, Chang CC, Lee YH, Cheng WT, et al. Amniotic fluid stem cells prevent follicle atresia and rescue fertility of mice with premature ovarian failure induced by chemotherapy. PLoS One. 2014;9:e106538. doi: 10.1371/journal.pone.0106538.
    1. Xiao GY, Cheng CC, Chiang YS, Cheng WT, Liu IH, Wu SC. Exosomal miR-10a derived from amniotic fluid stem cells preserves ovarian follicles after chemotherapy. Sci Rep. 2016;6:23120. doi: 10.1038/srep23120.
    1. Sun M, Wang S, Li Y, Yu L, Gu F, Wang C, et al. 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. Su J, Ding L, Cheng J, Yang J, Li X, Yan G, et al. 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. Wang S, Yu L, Sun M, Mu S, Wang C, Wang D, et al. The therapeutic potential of umbilical cord mesenchymal stem cells in mice premature ovarian failure. Biomed Res Int. 2013;2013:690491.
    1. Song D, Zhong Y, Qian C, Zou Q, Ou J, Shi Y, et al. Human umbilical cord mesenchymal stem cells therapy in cyclophosphamide-induced premature ovarian failure rat model. Biomed Res Int. 2016;2016:2517514.
    1. Liu T, Huang Y, Zhang J, Qin W, Chi H, Chen J, et al. Transplantation of human menstrual blood stem cells to treat premature ovarian failure in mouse model. Stem Cells Dev. 2014;23:1548–1557. doi: 10.1089/scd.2013.0371.
    1. Lai D, Wang F, Yao X, Zhang Q, Wu X, Xiang C. 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. Wang Z, Wang Y, Yang T, Li J, Yang X. Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice. Stem Cell Res Ther. 2017;8:11. doi: 10.1186/s13287-016-0458-1.
    1. Fairbairn NG, Randolph MA, Redmond RW. The clinical applications of human amnion in plastic surgery. J Plast Reconstr Aesthet Surg. 2014;67:662–675. doi: 10.1016/j.bjps.2014.01.031.
    1. Meesuk Ladda, Tantrawatpan Chairat, Kheolamai Pakpoom, Manochantr Sirikul. The immunosuppressive capacity of human mesenchymal stromal cells derived from amnion and bone marrow. Biochemistry and Biophysics Reports. 2016;8:34–40. doi: 10.1016/j.bbrep.2016.07.019.
    1. Ghasemzadeh M, Hosseini E, Ahmadi M, Kamalizad M, Amirizadeh N. Comparable osteogenic capacity of mesenchymal stem or stromal cells derived from human amnion membrane and bone marrow. Cytotechnology. 2018;70:729–739. doi: 10.1007/s10616-017-0177-1.
    1. Dabrowski FA, Burdzinska A, Kulesza A, Sladowska A, Zolocinska A, Gala K, et al. Comparison of the paracrine activity of mesenchymal stem cells derived from human umbilical cord, amniotic membrane and adipose tissue. J Obstet Gynaecol Res. 2017;43:1758–1768. doi: 10.1111/jog.13432.
    1. Manochantr S, U-pratya Y, Kheolamai P, Rojphisan S, Chayosumrit M, Tantrawatpan C, et al. Immunosuppressive properties of mesenchymal stromal cells derived from amnion, placenta, Wharton’s jelly and umbilical cord. Intern Med J. 2013;43:430–439. doi: 10.1111/imj.12044.
    1. Topoluk N, Hawkins R, Tokish J, Mercuri J. Amniotic mesenchymal stromal cells exhibit preferential osteogenic and chondrogenic differentiation and enhanced matrix production compared with adipose mesenchymal stromal cells. Am J Sports Med. 2017;45:2637–2646. doi: 10.1177/0363546517706138.
    1. Li Y, Liu Z, Jin Y, Zhu X, Wang S, Yang J, et al. Differentiation of human amniotic mesenchymal stem cells into human anterior cruciate ligament fibroblast cells by in vitro coculture. Biomed Res Int. 2017;2017:7360354.
    1. Wu Q, Fang T, Lang H, Chen M, Shi P, Pang X, et al. Comparison of the proliferation, migration and angiogenic properties of human amniotic epithelial and mesenchymal stem cells and their effects on endothelial cells. Int J Mol Med. 2017;39:918–926. doi: 10.3892/ijmm.2017.2897.
    1. Razavi Tousi SM, Faghihi M, Nobakht M, Molazem M, Kalantari E, Darbandi Azar A, et al. Improvement of heart failure by human amniotic mesenchymal stromal cell transplantation in rats. J Tehran Heart Cent. 2016;11:123–138.
    1. Kim SW, Zhang HZ, Kim CE, Kim JM, Kim MH. Amniotic mesenchymal stem cells with robust chemotactic properties are effective in the treatment of a myocardial infarction model. Int J Cardiol. 2013;168:1062–1069. doi: 10.1016/j.ijcard.2012.11.003.
    1. Li Y, Guo L, Ahn HS, Kim MH, Kim SW. Amniotic mesenchymal stem cells display neurovascular tropism and aid in the recovery of injured peripheral nerves. J Cell Mol Med. 2014;18:1028–1034. doi: 10.1111/jcmm.12249.
    1. Zhou H, Zhang H, Yan Z, Xu R. Transplantation of human amniotic mesenchymal stem cells promotes neurological recovery in an intracerebral hemorrhage rat model. Biochem Biophys Res Commun. 2016;475:202–208. doi: 10.1016/j.bbrc.2016.05.075.
    1. Jiao H, Shi K, Zhang W, Yang L, Yang L, Guan F, et al. Therapeutic potential of human amniotic membrane-derived mesenchymal stem cells in APP transgenic mice. Oncol Lett. 2016;12:1877–1883. doi: 10.3892/ol.2016.4857.
    1. Zhou HL, Zhang XJ, Zhang MY, Yan ZJ, Xu ZM, Xu RX. Transplantation of human amniotic mesenchymal stem cells promotes functional recovery in a rat model of traumatic spinal cord injury. Neurochem Res. 2016;41:2708–2718. doi: 10.1007/s11064-016-1987-9.
    1. Ding C, Li H, Wang Y, Wang F, Wu H, Chen R, et al. 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. Ling L, Feng X, Wei T, Wang Y, Wang Y, Zhang W, et al. Effects of low-intensity pulsed ultrasound (LIPUS)-pretreated human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation on primary ovarian insufficiency in rats. Stem Cell Res Ther. 2017;8:283. doi: 10.1186/s13287-017-0739-3.
    1. Qiang Y, Liang G, Yu L. Human amniotic mesenchymal stem cells alleviate lung injury induced by ischemia and reperfusion after cardiopulmonary bypass in dogs. Lab Investig. 2016;96:537–546. doi: 10.1038/labinvest.2016.37.
    1. Zhu L, Zhang YQ, Peng YL, Ren JJ. Effect of zuogui pill on GDF9/Smad2 signal transduction in premature ovarian failure model mice. Chin J Integr Med. 2017;37:1367–1371.
    1. Myers M, Britt KL, Wreford NG, Ebling FJ, Kerr JB. Methods for quantifying follicular numbers within the mouse ovary. Reproduction. 2004;127:569–580. doi: 10.1530/rep.1.00095.
    1. Xu SF, Yu LM, Fan ZH, Wu Q, Yuan Y, Wei Y, et al. Improvement of ginsenoside Rg1 on hematopoietic function in cyclophosphamide-induced myelosuppression mice. Eur J Pharmacol. 2012;695:7–12. doi: 10.1016/j.ejphar.2012.07.050.
    1. Yin N, Wang Y, Lu X, Liu R, Zhang L, Zhao W, 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;9:37. doi: 10.1186/s13287-018-0772-x.
    1. Rai P, Parrish M, Tay IJ, Li N, Ackerman S, He F, et al. Streptococcus pneumoniae secretes hydrogen peroxide leading to DNA damage and apoptosis in lung cells. Proc Natl Acad Sci U S A. 2015;112:E3421–E3430. doi: 10.1073/pnas.1424144112.
    1. van der Vliet A, Janssen-Heininger YM. Hydrogen peroxide as a damage signal in tissue injury and inflammation: murderer, mediator, or messenger? J Cell Biochem. 2014;115:427–435. doi: 10.1002/jcb.24683.
    1. Labunskyy VM, Gladyshev VN. Role of reactive oxygen species-mediated signaling in aging. Antioxid Redox Signal. 2013;19:1362–1372. doi: 10.1089/ars.2012.4891.
    1. Du X, Zhang L, Li X, Pan Z, Liu H, Li Q. TGF-β signaling controls FSHR signaling-reduced ovarian granulosa cell apoptosis through the SMAD4/miR-143 axis. Cell Death Dis. 2016;7:e2476. doi: 10.1038/cddis.2016.379.
    1. Schmidt D, Ovitt CE, Anlag K, Fehsenfeld S, Gredsted L, Treier AC, et al. The murine winged-helix transcription factor Foxl2 is required for granulosa cell differentiation and ovary maintenance. Development. 2004;131:933–942. doi: 10.1242/dev.00969.
    1. Pisarska MD, Barlow G, Kuo FT. Minireview: roles of the forkhead transcription factor FOXL2 in granulosa cell biology and pathology. Endocrinology. 2011;152:1199–1208. doi: 10.1210/en.2010-1041.
    1. Dai A, Sun H, Fang T, Zhang Q, Wu S, Jiang Y, et al. MicroRNA-133b stimulates ovarian estradiol synthesis by targeting Foxl2. FEBS Lett. 2013;587:2474–2482. doi: 10.1016/j.febslet.2013.06.023.
    1. Emori C, Sugiura K. Role of oocyte-derived paracrine factors in follicular development. Anim Sci J. 2014;85:627–633. doi: 10.1111/asj.12200.
    1. Cheng L, Sung MT, Cossu-Rocca P, Jones TD, MacLennan GT, De Jong J, et al. OCT4: biological functions and clinical applications as a marker of germ cell. J Pathol. 2007;211:1–9. doi: 10.1002/path.2105.
    1. Lee YM, Kim TH, Lee JH, Lee WJ, Jeon RH, Jang SJ, et al. Overexpression of Oct4 in porcine ovarian stem/stromal cells enhances differentiation of oocyte-like cells in vitro and ovarian follicular formation in vivo. J Ovarian Res. 2016;9:24. doi: 10.1186/s13048-016-0233-z.
    1. Paulini F, Melo EO. The role of oocyte-secreted factors GDF9 and BMP15 in follicular development and oogenesis. Reprod Domest Anim. 2011;46:354–361. doi: 10.1111/j.1439-0531.2010.01739.x.
    1. Orisaka M, Jiang JY, Orisaka S, Kotsuji F, Tsang BK. Growth differentiation factor 9 promotes rat preantral follicle growth by up-regulating follicular androgen biosynthesis. Endocrinology. 2009;150:2740–2748. doi: 10.1210/en.2008-1536.
    1. Li Y, Li RQ, Ou SB, Zhang NF, Ren L, Wei LN, et al. Increased GDF9 and BMP15 mRNA levels in cumulus granulosa cells correlate with oocyte maturation, fertilization, and embryo quality in humans. Reprod Biol Endocrinol. 2014;12:81. doi: 10.1186/1477-7827-12-81.
    1. Komatsu K, Koya T, Wang J, Yamashita M, Kikkawa F, Iwase A. Analysis of the effect of leukemia inhibitory factor on follicular growth in cultured murine ovarian tissue. Biol Reprod. 2015;93:18. doi: 10.1095/biolreprod.115.128421.
    1. Høyer PE, Byskov AG, Møllgård K. Stem cell factor and c-Kit in human primordial germ cells and fetal ovaries. Mol Cell Endocrinol. 2005;234:1–10. doi: 10.1016/j.mce.2004.09.012.
    1. Jin X, Han CS, Zhang XS, Yuan JX, Hu ZY, Liu YX. Signal transduction of stem cell factor in promoting early follicle development. Mol Cell Endocrinol. 2005;229:3–10. doi: 10.1016/j.mce.2004.10.006.

Source: PubMed

3
订阅