Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility

Qiuwan Zhang, Minhua Xu, Xiaofen Yao, Ting Li, Qian Wang, Dongmei Lai, Qiuwan Zhang, Minhua Xu, Xiaofen Yao, Ting Li, Qian Wang, Dongmei Lai

Abstract

Introduction: Premature ovarian failure and insufficiency are significant long-term side-effects of chemotherapy for female cancer patients. Recently, stem cell transplantation has been identified as a promising treatment for premature ovarian failure and insufficiency. We have previously demonstrated that human amniotic epithelial cells (hAECs) migrate into injured tissue and promote the recovery of ovarian function in chemoablated mice. However, the molecular mechanism guiding this process remains unclear.

Methods: To further investigate the effect of hAECs on chemotherapy-induced apoptosis, cultured primary hAECs were injected intravenously into mice treated with cyclophosphamide and busulphan. Apoptosis of granulosa cells was observed by TUNEL staining, and apoptosis-related gene expression was performed on ovarian tissue by real-time PCR and Western blot 7 days after hAEC transplantation. Additionally, the ovarian function and fertility of mice were assessed via counts of follicles and mating experiments at 4 weeks after hAEC transplantation.

Results: hAECs significantly inhibited tumor necrosis factor-alpha-mediated granulosa cell apoptosis induced by chemotherapeutics and reduced the inflammatory reaction in ovaries at 7 days after transplantation. In addition, 4 weeks after transplantation, hAECs promoted the development of follicles and increased the number of cumulus oocyte complexes in chemoablated mice. Furthermore, hAECs improved ovarian mass and increased the number of follicles compared to those of the chemoablated group, and hAEC transplantation partially rescued the fertility of chemoablated mice.

Conclusions: hAEC transplantation promotes ovarian function by inhibiting tumor necrosis factor-alpha-mediated cell apoptosis and reducing inflammation in chemotherapy-induced premature ovarian failure. These results suggest a potential molecular mechanism for the effective therapy of hAEC transplantation in chemotherapy-induced premature ovarian failure and insufficiency.

Figures

Fig. 1
Fig. 1
Chemotherapy reduced body weight of mice and the number of ovarian follicles. A Bar graph illustrating the body weight of mice in the sham and chemoablated (Cy) groups. B H&E staining of ovaries in sham and treatment groups, 7 and 10 days post-induction. Blue arrow indicated primordial follicle; red arrow indicated primary follicle; black arrow indicated secondary follicle; white arrow indicated mature follicle in B-b. C Bar graph representing the number of follicles at different stages of development. Data represent means ± SEM; *p < 0.05 versus Sham. Scale bar = 500 μm (a, c and d), 200 μm in b. Sham sham group (n = 6), Cy-3days chemoablated 3-day group (n = 6), Cy-7days chemoablated 7-day group (n = 6), Cy-10days chemoablated 10-day group (n = 6)
Fig. 2
Fig. 2
Chemotherapy-induced apoptosis of granulosa cells within developing follicle. A TUNEL-positive cells were observed in the ovary tissue at 7 days after chemotherapy. The red stain indicates TUNEL-positive granulosa cells. The blue DAPI stain indicates the cell nucleus. B Relative expression of Bcl2, Bax and TNF-α mRNA in ovarian tissue at 3, 7 and 10 days after chemotherapy. Data represent means ± SEM; *p < 0.05 versus Sham. Scale bar = 200 μm (a, b, c and d), 100 μm (e and f). Sham sham group (n = 6), Cy-3days chemoablated 3-day group (n = 6), Cy-7days chemoablated 7-day group (n = 6), Cy-10days chemoablated 10-day group (n = 6)
Fig. 3
Fig. 3
hAEC transplantation reduced chemotherapy-induced inflammation in ovarian tissue. A Section of dissected hAECs illustrated a cobble stone-like epithelial cell morphology. B Relative expression of mRNA levels detected by real-time PCR in cultured hAECs. C Image of fixed ovarian tissue in different groups at 7 days after hAEC transplantation. D Bar graph demonstrated the ovarian weights in different groups. E H&E staining results displaying ovarian morphology in different groups at 7 days after hAEC transplantation. F Relative expression of inflammatory cytokine mRNA levels in injured ovarian tissue analyzed by real-time PCR at 7 days after hAEC transplantation. Data represent means ± SEM; *p < 0.05 versus Sham, #p < 0.05 versus Cy. Scale bar = 200 μm in (E). Cy chemoablated group (n = 6), Cy + hAECs hAEC-treated group (n = 6), Sham sham group (n = 4)
Fig. 4
Fig. 4
hAEC transplantation inhibited chemotherapy-induced apoptosis in ovaries. A Relative mRNA expression of TNF-α, TRADD, FADD, caspase-3, Bcl2 and Bax was detected by real-time PCR in ovarian tissue at 7 days after hAECs transplantation. B Western blot results and corresponding densitometry analysis demonstrated that chemotherapy significantly increased activated caspase-3 protein expression at 7 days after hAEC transplantation. A significant reduction of activated caspase-3 protein expression in the hAEC-treated group compared with the chemoablated group. Data are mean ± SEM; *p < 0.05 versus Sham; #p < 0.05 versus Cy. Cy chemoablated group (n = 6 in A, n = 4 in B), Cy + hAECs hAEC-treated group (n = 6 in A, n = 4 in B), Sham sham group (n = 4 in both panels)
Fig. 5
Fig. 5
hAEC transplantation increased the number of cumulus oocyte complexes (COC) in chemoablated mice. AC Images of ovarian morphology followed superovulation in different groups 1 month after hAEC transplantation (white arrows). DF The morphology of COC was observed under microscopy. G Bar graph representing the counting of COC in different groups. HJ H&E staining of ovaries followed superovulation was used to observed follicle development. Enlargements of outlined areas are shown in panels af. Mature follicles are indicated by the black arrow (a and e). Primary follicles are indicated by the black arrowhead (b, d and f). Follicular atresia is indicated by the white arrowhead (c). Data are mean ± SEM; *p < 0.05 versus Sham; #p < 0.05 versus Cy. Scale bars = 200 μm (HJ), 100 μm (a, c and e), 50 μm (b, d and f). Cy chemoablated group (n = 4), Cy + hAECs hAEC-treated group (n = 4), Sham sham group (n = 4)
Fig. 6
Fig. 6
hAEC transplantation increased the number of ovarian follicles and restored the fertility of chemoablated mice. A Bar graph illustrating body weight of experimental and control groups following hAEC transplantation. B The ovarian weight in different groups at 28 days after hAEC transplantation. C Histological analysis of ovaries in different groups at 28 days after hAEC transplantation. D The number of different stage follicles counted at 28 days after hAEC transplantation. E Bar graph representing the number of pups per pregnancy at the end of mating. Data are mean ± SEM; *p < 0.05 versus Sham; #p < 0.05 versus Cy. Scale bars in c = 500 μm (a–c), 200 μm (d–f), 100 μm (g–i), 50 μm (j–l). Cy chemoablated group (n = 6; n = 10 in E), Cy + hAECs hAEC-treated group (n = 6; n = 10 in E), Sham sham group (n = 6; n = 5 in E)

References

    1. Goswami D, Conway GS. Premature ovarian failure. Hum Reprod Update. 2005;11:391–410. doi: 10.1093/humupd/dmi012.
    1. Sadeghi MR. New hopes for the treatment of primary ovarian insufficiency/premature ovarian failure. J Reprod Infertil. 2013;14:1–2.
    1. Panay N, Fenton A. Premature ovarian failure: a growing concern. Climacteric. 2008;11:1–3. doi: 10.1080/13697130701878635.
    1. White YA, Woods DC, Takai Y, Ishihara O, Seki H, Tilly JL. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat Med. 2012;18:413–21. doi: 10.1038/nm.2669.
    1. Lai D, Wang F, Dong Z, Zhang Q. Skin-derived mesenchymal stem cells help restore function to ovaries in a premature ovarian failure mouse model. PLoS One. 2014;9:e98749. doi: 10.1371/journal.pone.0098749.
    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. 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. Whittle WL, Gibb W, Challis JR. The characterization of human amnion epithelial and mesenchymal cells: the cellular expression, activity and glucocorticoid regulation of prostaglandin output. Placenta. 2000;21:394–401. doi: 10.1053/plac.1999.0482.
    1. Miki T, Lehmann T, Cai H, Stolz DB, Strom SC. Stem cell characteristics of amniotic epithelial cells. Stem Cells. 2005;23:1549–59. doi: 10.1634/stemcells.2004-0357.
    1. Ilancheran S, Michalska A, Peh G, Wallace EM, Pera M, Manuelpillai U. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biol Reprod. 2007;77:577–88. doi: 10.1095/biolreprod.106.055244.
    1. Vosdoganes P, Lim R, Koulaeva E, Chan ST, Acharya R, Moss TJ, et al. Human amnion epithelial cells modulate hyperoxia-induced neonatal lung injury in mice. Cytotherapy. 2013;15:1021–9. doi: 10.1016/j.jcyt.2013.03.004.
    1. Vosdoganes P, Wallace EM, Chan ST, Acharya R, Moss TJ, Lim R. Human amnion epithelial cells repair established lung injury. Cell Transplant. 2013;22:1337–49. doi: 10.3727/096368912X657657.
    1. Tan JL, Chan ST, Wallace EM, Lim R. Human amnion epithelial cells mediate lung repair by directly modulating macrophage recruitment and polarization. Cell Transplant. 2014;23:319–28. doi: 10.3727/096368912X661409.
    1. Yawno T, Schuilwerve J, Moss TJ, Vosdoganes P, Westover AJ, Afandi E, et al. Human amnion epithelial cells reduce fetal brain injury in response to intrauterine inflammation. Dev Neurosci. 2013;35:272–82. doi: 10.1159/000346683.
    1. Lim R, Chan ST, Tan JL, Mockler JC, Murphy SV, Wallace EM. Preterm human amnion epithelial cells have limited reparative potential. Placenta. 2013;34:486–92. doi: 10.1016/j.placenta.2013.03.010.
    1. Evron A, Goldman S, Shalev E. Human amniotic epithelial cells differentiate into cells expressing germ cell specific markers when cultured in medium containing serum substitute supplement. Reprod Biol Endocrinol. 2012;10:108. doi: 10.1186/1477-7827-10-108.
    1. Wang F, Wang L, Yao X, Lai D, Guo L. Human amniotic epithelial cells can differentiate into granulosa cells and restore folliculogenesis in a mouse model of chemotherapy-induced premature ovarian failure. Stem Cell Res Ther. 2013;4:124. doi: 10.1186/scrt335.
    1. Pedersen T, Peters H. Proposal for a classification of oocytes and follicles in the mouse ovary. J Reprod Fertil. 1968;17:555–7. doi: 10.1530/jrf.0.0170555.
    1. Kalich-Philosoph L, Roness H, Carmely A, Fishel-Bartal M, Ligumsky H, Paglin S, et al. Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Sci Transl Med. 2013;5:185ra62. doi: 10.1126/scitranslmed.3005402.
    1. Aikawa NE, Sallum AM, Pereira RM, Suzuki L, Viana VS, Bonfa E, et al. Subclinical impairment of ovarian reserve in juvenile systemic lupus erythematosus after cyclophosphamide therapy. Clin Exp Rheumatol. 2012;30:445–9.
    1. Greenfeld CR, Roby KF, Pepling ME, Babus JK, Terranova PF, Flaws JA. Tumor necrosis factor (TNF) receptor type 2 is an important mediator of TNF alpha function in the mouse ovary. Biol Reprod. 2007;76:224–31. doi: 10.1095/biolreprod.106.055509.
    1. Geering B, Gurzeler U, Federzoni E, Kaufmann T, Simon HU. A novel TNFR1-triggered apoptosis pathway mediated by class IA PI3Ks in neutrophils. Blood. 2011;117:5953–62. doi: 10.1182/blood-2010-11-322206.
    1. Gonfloni S, Di Tella L, Caldarola S, Cannata SM, Klinger FG, Di Bartolomeo C, et al. Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death. Nat Med. 2009;15:1179–85. doi: 10.1038/nm.2033.
    1. Markstrom E, Svensson E, Shao R, Svanberg B, Billig H. Survival factors regulating ovarian apoptosis—dependence on follicle differentiation. Reproduction. 2002;123:23–30. doi: 10.1530/rep.0.1230023.
    1. Tan SJ, Lee LJ, Tzeng CR, Wang CW, Hsu MI, Chen CH. Targeted anti-apoptosis activity for ovarian protection against chemotherapy-induced ovarian gonadotoxicity. Reprod Biomed Online. 2014. doi:10.1016/j.rbmo.2014.07.014.
    1. Lopes F, Smith R, Anderson RA, Spears N. Docetaxel induces moderate ovarian toxicity in mice, primarily affecting granulosa cells of early growing follicles. Mol Hum Reprod. 2014;20:948–59. doi: 10.1093/molehr/gau057.
    1. Hutt KJ. The role of BH3-only proteins in apoptosis within the ovary. Reproduction. 2015;149:R81–9. doi: 10.1530/REP-14-0422.
    1. Ganz PA, Bower JE, Kwan L, Castellon SA, Silverman DH, Geist C, et al. Does tumor necrosis factor-alpha (TNF-alpha) play a role in post-chemotherapy cerebral dysfunction? Brain Behav Immun. 2013;30:S99–108. doi: 10.1016/j.bbi.2012.07.015.
    1. Park MR, Choi YJ, Kwon DN, Park C, Bui HT, Gurunathan S, et al. Intraovarian transplantation of primordial follicles fails to rescue chemotherapy injured ovaries. Sci Rep. 2013;3:1384.
    1. Diaz-Prado S, Muinos-Lopez E, Hermida-Gomez T, Rendal-Vazquez ME, Fuentes-Boquete I, de Toro FJ, et al. Multilineage differentiation potential of cells isolated from the human amniotic membrane. J Cell Biochem. 2010;111:846–57. doi: 10.1002/jcb.22769.
    1. Hou Y, Huang Q, Liu T, Guo L. Human amnion epithelial cells can be induced to differentiate into functional insulin-producing cells. Acta Biochim Biophys Sin. 2008;40:830–9. doi: 10.1093/abbs/40.9.830.
    1. Jones CA, Williams KA, Finlay-Jones JJ, Hart PH. Interleukin 4 production by human amnion epithelial cells and regulation of its activity by glycosaminoglycan binding. Biol Reprod. 1995;52:839–47. doi: 10.1095/biolreprod52.4.839.
    1. Murphy SV, Shiyun SC, Tan JL, Chan S, Jenkin G, Wallace EM, et al. Human amnion epithelial cells do not abrogate pulmonary fibrosis in mice with impaired macrophage function. Cell Transplant. 2012;21:1477–92. doi: 10.3727/096368911X601028.
    1. Grzywocz Z, Pius-Sadowska E, Klos P, Gryzik M, Wasilewska D, Aleksandrowicz B, et al. Growth factors and their receptors derived from human amniotic cells in vitro. Folia Histochem Cytobiol. 2014;52:163–70. doi: 10.5603/FHC.2014.0019.

Source: PubMed

3
Subskrybuj