Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure

Min Sun, Shufang Wang, Yi Li, Ling Yu, Fang Gu, Changyong Wang, Yuanqing Yao, Min Sun, Shufang Wang, Yi Li, Ling Yu, Fang Gu, Changyong Wang, Yuanqing Yao

Abstract

Introduction: Young patients receiving chemotherapy occasionally face infertility and premature ovarian failure (POF). Numerous investigations reported that adipose-derived stem cells (ADSCs) transplantation could ameliorate the structure and function of injured tissues. The aim of this study was to explore the therapeutic efficacy of ADSC transplantation for chemotherapy-induced ovarian damage.

Methods: Female mice were injected intraperitoneally with 50 mg/kg cyclophosphamide (CTX). After 15 consecutive days of injection, ADSCs were transplanted either directly into bilateral ovaries or via intravenous injection, and the ovaries were excised after either 1 week or 1 month of treatment. The follicles were counted and categorized, and ovarian histologic sections were stained for TUNEL. Ovarian function was evaluated by monitoring ovulation. ADSC tracking, microarray analyses, and real-time polymerase chain reaction (PCR) were used to assess the inner mechanism of injury and repair.

Results: The ovarian function of mice exposed to CTX injection improved after ADSC transplantation. The population of follicles at different stages and ovulation significantly increased after the treatment. Immunofluorescence revealed reduced TUNEL staining. The tracking of ADSCs revealed that these cells did not directly differentiate into the follicle component. Microarray analyses indicated that changes in different groups of genes might affect follicle formation or ovulation.

Conclusions: ADSC transplantation improved ovarian function. Our results suggest a potential mechanism for ADSC therapy.

Figures

Figure 1
Figure 1
The isolation and identification of ADSCs. (A) The ADSCs exhibited typical fibroblastic morphology. (B) FCM analysis of ADSCs. The cells were positive for CD29, CD44, and CD90 and negative for CD31, CD34, and CD45. (C) ADSCs differentiate into osteoblasts and adipocytes. Scale bars: 100 μm.
Figure 2
Figure 2
The follicle number increased after transplantation. (A) Hematoxylin and eosin staining. (a) Most of the follicles of WT and ADSCs were secondary and antral follicles. The population of follicles decreased significantly after injection with CTX (b) and recovered after the ADSCs both intravenous and in situ therapy (c, d). The POF model mouse had fewer follicles and most of them were primary and secondary follicles or had no function. (B) The number of follicles after one week. (C) The number of follicles after 1 month. Scale bars: 200 μm. (*P < 0.05 in situ versus POF group; #P < 0.05 intravenous versus POF group).
Figure 3
Figure 3
The ovulation increased after therapy. (A) The oocytes of the WT group. (B) The oocytes in the POF model exhibited abnormal morphology and number. (C) The oocytes of the intravenous group. (D) The oocytes of the in situ group. (E, F) The population of oocytes increased after 1 week and 1 month of ADSC therapy. (*P < 0.05 intravenous versus the POF group, #P < 0.05 in situ versus the POF group).
Figure 4
Figure 4
The apoptosis of ovaries reduced after ADSC therapy. The ovaries of WT group (A, E), POF group (B, F), the intravenous group (C, G), and the in situ group (D, H). The green stain indicates TUNEL-positive GCs. The blue stain indicates the nucleus. Data from one week demonstrate that apoptosis was attenuated by the transplantation of ADSCs. (I) The number of apoptotic GCs decreased after 1 week of ADSC therapy. Scale bars: 100 μm (*P < 0.05 intravenous versus POF group, #P < 0.05 in situ versus POF group).
Figure 5
Figure 5
ADSCs tracking in vivo. (A) GFP (+)-ADSCs under a light microscope. (B) GFP (+)-ADSCs under a fluorescence microscope. (C, E) One week and 1 month after transplantation, ADSCs can be traced in the intravenous group. (D, F) One week and 1 month after transplantation, ADSCs can be traced in the in situ group. (G) ADSCs can not be traced in WT group. Scale bars: 100 μm.
Figure 6
Figure 6
Transplanted ADSCs were not formed into oocytes. (A) Oocytes and cumulus cells from donor ovaries showed green fluorescence. (B, C) Oocytes and cumulus cells from recipient ovaries did not show green fluorescence. Scale bars: 100 μm.
Figure 7
Figure 7
Microarray and real-time PCR analysis. (A) Gene expression in the intravenous group versus the WT group. (B) Gene expression in the intravenous group versus the WT group. (C) Gene expression in the in situ group versus the WT group. (D) Relative expression of genes. Angpt1, Onecut2, and Zcchc11 were upregulated, and Cxcr4 was downregulated; these results were in accordance with the microarray result.

References

    1. Hoyer PB, Sipes IG. Assessment of follicle destruction in chemical-induced ovarian toxicity. Annu Rev Pharmacol Toxicol. 1996;36:307–331. doi: 10.1146/annurev.pa.36.040196.001515.
    1. Anasti JN. Premature ovarian failure: an update. Fertil Steril. 1998;70:1–15.
    1. Molina JR, Barton DL, Loprinzi CL. Chemotherapy-induced ovarian failure: manifestations and management. Drug Saf. 2005;28:401–416. doi: 10.2165/00002018-200528050-00004.
    1. De Vos M, Devroey P, Fauser BC. Primary ovarian insufficiency. Lancet. 2010;376:911–921. doi: 10.1016/S0140-6736(10)60355-8.
    1. Yucebilgin MS, Terek MC, Ozsaran A, Akercan F, Zekioglu O, Isik E, Erhan Y. Effect of chemotherapy on primordial follicular reserve of rat: an animal model of premature ovarian failure and infertility. Aust N Z J Obstet Gynaecol. 2004;44:6–9. doi: 10.1111/j.1479-828X.2004.00143.x.
    1. Fleischer RT, Vollenhoven BJ, Weston GC. The effects of chemotherapy and radiotherapy on fertility in premenopausal women. Obstet Gynecol Surv. 2011;66:248–254. doi: 10.1097/OGX.0b013e318224e97b.
    1. Lee HJ, Selesniemi K, Niikura Y, Niikura T, Klein R, Dombkowski DM, Tilly JL. Bone marrow transplantation generates immature oocytes and rescues long-term fertility in a preclinical mouse model of chemotherapy-induced premature ovarian failure. J Clin Oncol. 2007;25:3198–3204. doi: 10.1200/JCO.2006.10.3028.
    1. Fu X, He Y, Xie C, Liu W. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy. 2008;10:353–363. doi: 10.1080/14653240802035926.
    1. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–228. doi: 10.1089/107632701300062859.
    1. Varma MJ, Breuls RG, Schouten TE, Jurgens WJ, Bontkes HJ, Schuurhuis GJ, van Ham SM, van Milligen FJ. Phenotypical and functional characterization of freshly isolated adipose tissue-derived stem cells. Stem Cells Dev. 2007;16:91–104. doi: 10.1089/scd.2006.0026.
    1. Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K, Bae YC, Jung JS. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem. 2004;14:311–324. doi: 10.1159/000080341.
    1. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–4295. doi: 10.1091/mbc.E02-02-0105.
    1. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100:1249–1260. doi: 10.1161/01.RES.0000265074.83288.09.
    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. Fujimura J, Ogawa R, Mizuno H, Fukunaga Y, Suzuki H. Neural differentiation of adipose-derived stem cells isolated from GFP transgenic mice. Biochem Biophys Res Commun. 2005;333:116–121. doi: 10.1016/j.bbrc.2005.05.096.
    1. Lopez SG, Luderer U. Effects of cyclophosphamide and buthionine sulfoximine on ovarian glutathione and apoptosis. Free Radic Biol Med. 2004;36:1366–1377. doi: 10.1016/j.freeradbiomed.2004.02.067.
    1. Rehman J, Traktuev D, Li J, Merfeld-Clauss S, Temm-Grove CJ, Bovenkerk JE, Pell CL, Johnstone BH, Considine RV, March KL. Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells. Circulation. 2004;109:1292–1298. doi: 10.1161/01.CIR.0000121425.42966.F1.
    1. Nakagami H, Maeda K, Morishita R, Iguchi S, Nishikawa T, Takami Y, Kikuchi Y, Saito Y, Tamai K, Ogihara T, Kaneda Y. Novel autologous cell therapy in ischemic limb disease through growth factor secretion by cultured adipose tissue-derived stromal cells. Arterioscler Thromb Vasc Biol. 2005;25:2542–2547. doi: 10.1161/01.ATV.0000190701.92007.6d.
    1. Nakagami H, Morishita R, Maeda K, Kikuchi Y, Ogihara T, Kaneda Y. Adipose tissue-derived stromal cells as a novel option for regenerative cell therapy. J Atheroscler Thromb. 2006;13:77–81. doi: 10.5551/jat.13.77.
    1. Cao Y, Sun Z, Liao L, Meng Y, Han Q, Zhao RC. Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo. Biochem Biophys Res Commun. 2005;332:370–379. doi: 10.1016/j.bbrc.2005.04.135.
    1. Prueitt RL, Zinn AR. A fork in the road to fertility. Nat Genet. 2001;27:132–134. doi: 10.1038/84735.
    1. Nishigaki A, Okada H, Tsuzuki T, Cho H, Yasuda K, Kanzaki H. Angiopoietin 1 and angiopoietin 2 in follicular fluid of women undergoing a long protocol. Fertil Steril. 2011;96:1378–1383. doi: 10.1016/j.fertnstert.2011.09.031.
    1. Holt JE, Jackson A, Roman SD, Aitken RJ, Koopman P, McLaughlin EA. CXCR4/SDF1 interaction inhibits the primordial to primary follicle transition in the neonatal mouse ovary. Dev Biol. 2006;293:449–460. doi: 10.1016/j.ydbio.2006.02.012.
    1. Jones MR, Blahna MT, Kozlowski E, Matsuura KY, Ferrari JD, Morris SA, Powers JT, Daley GQ, Quinton LJ, Mizgerd JP. Zcchc11 uridylates mature miRNAs to enhance neonatal IGF-1 expression, growth, and survival. PLoS Genet. 2012;8:e1003105. doi: 10.1371/journal.pgen.1003105.
    1. Mao J, Smith MF, Rucker EB, Wu GM, McCauley TC, Cantley TC, Prather RS, Didion BA, Day BN. Effect of epidermal growth factor and insulin-like growth factor I on porcine preantral follicular growth, antrum formation, and stimulation of granulosal cell proliferation and suppression of apoptosis in vitro. J Anim Sci. 2004;82:1967–1975.
    1. Sirotkin AV, Dukesova J, Pivko J, Makarevich AV, Kubek A. Effect of growth factors on proliferation, apoptosis and protein kinase A expression in cultured porcine cumulus oophorus cells. Reprod Nutr Dev. 2002;42:35–43. doi: 10.1051/rnd:2002004.
    1. Mazerbourg S, Bondy CA, Zhou J, Monget P. The insulin-like growth factor system: a key determinant role in the growth and selection of ovarian follicles? A comparative species study. Reprod Domest Anim. 2003;38:247–258. doi: 10.1046/j.1439-0531.2003.00440.x.
    1. Tilly JL. Commuting the death sentence: how oocytes strive to survive. Nat Rev Mol Cell Biol. 2001;2:838–848. doi: 10.1038/35099086.
    1. Kucerova L, Matuskova M, Pastorakova A, Tyciakova S, Jakubikova J, Bohovic R, Altanerova V, Altaner C. Cytosine deaminase expressing human mesenchymal stem cells mediated tumour regression in melanoma bearing mice. J Gene Med. 2008;10:1071–1082. doi: 10.1002/jgm.1239.

Source: PubMed

3
購読する