DOI QR코드

DOI QR Code

Homing and Restorative Effects of Bone Marrow-Derived Mesenchymal Stem Cells on Cisplatin Injured Ovaries in Rats

  • Liu, Jiabin (Liaoning Medical University) ;
  • Zhang, Haiying (Liaoning Medical University) ;
  • Zhang, Yun (Department of Gynecology and Obstetrics, Third Affiliated Hospital of Liaoning Medical University) ;
  • Li, Nan (Department of Gynecology and Obstetrics, Third Affiliated Hospital of Liaoning Medical University) ;
  • Wen, Yuku (Department of Gynecology and Obstetrics, Third Affiliated Hospital of Liaoning Medical University) ;
  • Cao, Fanglei (Department of Gynecology and Obstetrics, Third Affiliated Hospital of Liaoning Medical University) ;
  • Ai, Hao (Liaoning Medical University) ;
  • Xue, Xiaoou (Dongzhimen Hospital Affiliated to Beijing University of China Medicine)
  • Received : 2014.06.02
  • Accepted : 2014.09.23
  • Published : 2014.12.31

Abstract

Premature ovarian failure (POF) is a long-term adverse effect of chemotherapy treatment. However, current available treatment regimens are not optimal. Emerging evidence suggests that bone marrow-derived mesenchymal stem cells (BMSCs) could restore the structure and function of injured tissues, but the homing and restorative effects of BMSCs on chemotherapy injured ovaries are still not clear. In this study, we found that granulosa cell (GC) apoptosis induced by cisplatin was reduced when BMSCs were migrated to granulosa cells (GCs) in vitro. Chemotherapy-induced POF was induced by intraperitoneal injection of cisplatin in rats. BMSCs labeled with enhanced green fluorescent protein (EGFP) were injected into the rats via the tail vein to investigate the homing and distribution of BMSCs in vivo. The number of BMSCs in the ovarian hilum and medulla was greater than in the cortex, but no BMSCs were found in the follicles and corpus lutea. In addition, the BMSCs treatment group's antral follicle count and estradiol levels increased after 30 days, compared with the POF group. Hence, our study demonstrates that intravenously delivered BMSCs can home to the ovaries, and restore its structure and function in POF model rats.

Keywords

References

  1. Abd-Allah, S.H., Shalaby, S.M., Pasha, H.F., El-Shal, A.S., Raafat, N., Shabrawy, S.M., Awad, H.A., Amer, M.G., Gharib, M.A., El Gendy, E.A., et al. (2013). Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy 15, 64-75. https://doi.org/10.1016/j.jcyt.2012.08.001
  2. Agung, M., Ochi, M., Yanada, S., Adachi, N., Izuta, Y., Yamasaki, T., and Toda, K. (2006). Mobilization of bone marrow-derived mesenchymal stem cells into the injured tissues after intraarticular injection and their contribution to tissue regeneration. Knee Surg. Sports Traumatol. Arthrosc. 14, 1307-1314. https://doi.org/10.1007/s00167-006-0124-8
  3. Ai, H., Zhang, H., Zhang, Y., and Liu, Y. (2012). Mouse ovarian-related gene expression profiles change with intraperitoneal injection of cisplatin. African J. Pharm. Pharm. 6, 2119-2122.
  4. Antoine, C., Ameye, L., Paesmans, M., and Rozenberg, S. (2013). Systematic review about breast cancer incidence in relation to hormone replacement therapy use. Climacteric 17, 116-135.
  5. Bi, B., Schmitt, R., Israilova, M., Nishio, H., and Cantley, L.G. (2007). Stromal cells protect against acute tubular injury via an endocrine effect. J. Am. Soc. Nephrol.18, 2486-2496. https://doi.org/10.1681/ASN.2007020140
  6. Bosch, P., Fouletier-Dilling, C., Olmsted-Davis, E., Davis, A., and Stice, S. (2006). Efficient adenoviral-mediated gene delivery into porcine mesenchymal stem cells. Mol. Reprod. Dev. 73, 1393-1403. https://doi.org/10.1002/mrd.20593
  7. Das, B., Antoon, R., Tsuchida, R., Lotfi, S., Morozova, O., Farhat, W., Malkin, D., Koren, G., Yeger, H., and Baruchel, S. (2008). Squalene selectively protects mouse bone marrow progenitors against cisplatin and carboplatin-induced cytotoxicity in vivo without protecting tumor growth. Neoplasia 10, 1105-1119. https://doi.org/10.1593/neo.08466
  8. De Vos, F., Nuver, J., Willemse, P., van der Zee, A., Messerschmidt, J., Burgerhof, J., De Vries, E., and Gietema, J. (2004). Long-term survivors of ovarian malignancies after cisplatin-based chemotherapy: cardiovascular risk factors and signs of vascular damage. Eur. J. Cancer 40, 696-700.
  9. Donnez, J., and Dolmans, M.-M. (2013). Fertility preservation in women. Nat. Rev. Endocrinol. 9, 735-749. https://doi.org/10.1038/nrendo.2013.205
  10. Forte, G., Minieri, M., Cossa, P., Antenucci, D., Sala, M., Gnocchi, V., Fiaccavento, R., Carotenuto, F., De Vito, P., Baldini, P.M., et al. (2006). Hepatocyte growth factor effects on mesenchymal stem cells: proliferation, migration, and differentiation. Stem Cells 24, 23-33. https://doi.org/10.1634/stemcells.2004-0176
  11. Frenette, P.S., Pinho, S., Lucas, D., and Scheiermann, C. (2013). Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Ann. Rev. Immunol. 31, 285-316. https://doi.org/10.1146/annurev-immunol-032712-095919
  12. Fu, X., He, Y., Xie, C., and Liu, W. (2008). Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy 10, 353-363. https://doi.org/10.1080/14653240802035926
  13. Ghadami, M., El-Demerdash, E., Zhang, D., Salama, S.A., Binhazim, A.A., Archibong, A.E., Chen, X., Ballard, B.R., Sairam, M.R., and Al-Hendy, A. (2012). Bone marrow transplantation restores follicular maturation and steroid hormones production in a mouse model for primary ovarian failure. PLoS One 7, e32462. https://doi.org/10.1371/journal.pone.0032462
  14. Guo, J.Q., Gao, X., Lin, Z.J., Wu, W.Z., Huang, L.H., Dong, H.Y., Chen, J., Lu, J., Fu, Y.F., Wang, J., et al. (2013). BMSCs reduce rat granulosa cell apoptosis induced by cisplatin and perimenopause. BMC Cell Biol. 14, 1-9. https://doi.org/10.1186/1471-2121-14-1
  15. Iorio, R., Castellucci, A., Ventriglia, G., Teoli, F., Cellini, V., Macchiarelli, G., and Cecconi, S. (2014). Ovarian toxicity: from environmental exposure to chemotherapy. Curr. Pharm. Des. 20, 5388-5397. https://doi.org/10.2174/1381612820666140205145319
  16. Johnson, J., Bagley, J., Skaznik-Wikiel, M., Lee, H.-J., Adams, G.B., Niikura, Y., Tschudy, K.S., Tilly, J.C., Cortes, M.L., Forkert, R., et al. (2005). Oocyte generation in adult mammalian ovaries by putative germ cells in bone marrow and peripheral blood. Cell 122, 303-315. https://doi.org/10.1016/j.cell.2005.06.031
  17. Kilic, S., Pinarli, F., Ozogul, C., Tasdemir, N., Naz Sarac, G., and Delibasi, T. (2013). Protection from cyclophosphamide-induced ovarian damage with bone marrow-derived mesenchymal stem cells during puberty. Gynecol. Endocrinol. 30, 135-140.
  18. Kupcova Skalnikova, H. (2013). Proteomic techniques for characterisation of mesenchymal stem cell secretome. Biochimie 95, 2196-2211. https://doi.org/10.1016/j.biochi.2013.07.015
  19. Lai, D., Wang, F., Chen, Y., Wang, L., Wang, Y., and Cheng, W. (2013). Human amniotic fluid stem cells have a potential to recover ovarian function in mice with chemotherapy-induced sterility. BMC Dev. Biol. 13, 34. https://doi.org/10.1186/1471-213X-13-34
  20. Lana, M.B.P., Straminsky, V., Onetto, C., Amuchastegui, J.M., Blanco, G., Galluzzo, L., Provenzano, S., and Nolting, M. (2010). What is really responsible for bone loss in spontaneous premature ovarian failure? A new enigma. Gynecol. Endocrinol. 26, 755-759. https://doi.org/10.3109/09513590.2010.487599
  21. Lee, O., Coathup, M., Goodship, A., and Blunn, G. (2005). Use of mesenchymal stem cells to facilitate bone regeneration in normal and chemotherapy-treated rats. Tissue Eng. 11, 1727-1735. https://doi.org/10.1089/ten.2005.11.1727
  22. Lee, H.-J., Selesniemi, K., Niikura, Y., Niikura, T., Klein, R., Dombkowski, D.M., and Tilly, J.L. (2007). 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. 25, 3198-3204. https://doi.org/10.1200/JCO.2006.10.3028
  23. Liu, T., Huang, Y., Guo, L., Cheng, W., and Zou, G. (2012). 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. 9, 592-602. https://doi.org/10.7150/ijms.4841
  24. Lue, Y., Erkkila, K., Liu, P.Y., Ma, K., Wang, C., Hikim, A.S., and Swerdloff, R.S. (2007). Fate of bone marrow stem cells transplanted into the testis: potential implication for men with testicular failure. Am. J. Pathol. 170, 899-908. https://doi.org/10.2353/ajpath.2007.060543
  25. Morse, H., Elfving, M., Lindgren, A., Wolner-Hanssen, P., Andersen, C.Y., and Ora, I. (2013). Acute onset of ovarian dysfunction in young females after start of cancer treatment. Pediatr. Blood Cancer 60, 676-681. https://doi.org/10.1002/pbc.24327
  26. Maclaran, K., and Panay, N. (2011). Premature ovarian failure. J. Fam. Plann. Reprod. Health Care 37, 35-42. https://doi.org/10.1136/jfprhc.2010.0015
  27. Maruo, T., Laoag-Fernandez, J., Takekida, S., Peng, X., Deguchi, J., Samoto, T., Kondo, H., and Matsuo, H. (1999). Regulation of granulosa cell proliferation and apoptosis during follicular development. Gynecol. Endocrinol. 13, 410-419. https://doi.org/10.3109/09513599909167588
  28. Matsuda, F., Inoue, N., Manabe, N., and Ohkura, S. (2011). Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J. Reprod. Dev. 58, 44-50.
  29. Miyamoto, Y., Shimada, K., Sakaguchi, Y., and Miyamoto, M. (2007). Cisplatin (CDDP)-induced acute toxicity in an experimental model of hepatic fibrosis. J. Toxicol. Sci. 32, 311-319. https://doi.org/10.2131/jts.32.311
  30. Muscari, C., Bonafe, F., Martin-Suarez, S., Valgimigli, S., Valente, S., Fiumana, E., Fiorelli, F., Rubini, G., Guarnieri, C., Caldarera, C.M., et al. (2013). Restored perfusion and reduced inflammation in the infarcted heart after grafting stem cells with a hyaluronan-based scaffold. J. Cell. Mol. Med. 17, 518-530. https://doi.org/10.1111/jcmm.12039
  31. Nayernia, K., Lee, J.H., Drusenheimer, N., Nolte, J., Wulf, G., Dressel, R., Gromoll, J., and Engel, W. (2006). Derivation of male germ cells from bone marrow stem cells. Lab. Invest. 86, 654-663. https://doi.org/10.1038/labinvest.3700429
  32. Nozaki, Y., Furubo, E., Matsuno, T., Fukui, R., Kizawa, K., Kozaki, T., and Sanzen, T. (2009). Collaborative work on evaluation of ovarian toxicity 6). Two-or four-week repeated-dose studies and fertility study of cisplatin in female rats. J. Toxicol. Sci. 34, SP73-81.
  33. Ohishi, M., and Schipani, E. (2010). Bone marrow mesenchymal stem cells. J. Cell. Biochem. 109, 277-282.
  34. Ruster, B., Grace, B., Seitz, O., Seifried, E., and Henschler, R. (2005). Induction and detection of human mesenchymal stem cell migration in the 48-well reusable transwell assay. Stem Cells Dev. 14, 231-235. https://doi.org/10.1089/scd.2005.14.231
  35. Santos, N., Catao, C., Martins, N., Curti, C., Bianchi, M., and Santos, A. (2007). Cisplatin-induced nephrotoxicity is associated with oxidative stress, redox state unbalance, impairment of energetic metabolism and apoptosis in rat kidney mitochondria. Arch. Toxicol. 81, 495-504. https://doi.org/10.1007/s00204-006-0173-2
  36. Schmidt, A., Ladage, D., Schinkothe, T., Klausmann, U., Ulrichs, C., Klinz, F.J., Brixius, K., Arnhold, S., Desai, B., and Mehlhorn, U. (2006). Basic fibroblast growth factor controls migration in human mesenchymal stem cells. Stem Cells 24, 1750-1758. https://doi.org/10.1634/stemcells.2005-0191
  37. Selesniemi, K., Lee, H.-J., Niikura, T., and Tilly, J.L. (2009). Young adult donor bone marrow infusions into female mice postpone age-related reproductive failure and improve offspring survival. Aging 1, 49-57.
  38. Siddik, Z.H. (2003). Cisplatin: mode of cytotoxic action and molecular basis of resistance. Oncogene 22, 7265-7279. https://doi.org/10.1038/sj.onc.1206933
  39. Sohni, A., and Verfaillie, C.M. (2013). Mesenchymal stem cells migration homing and tracking. Stem Cells Int. 2013, 130763.
  40. Takehara, Y., Yabuuchi, A., Ezoe, K., Kuroda, T., Yamadera, R., Sano, C., Murata, N., Aida, T., Nakama, K., Aono, F., et al. (2013). The restorative effects of adipose-derived mesenchymal stem cells on damaged ovarian function. Lab. Invest. 93, 181-193. https://doi.org/10.1038/labinvest.2012.167
  41. Tannock, I.F., Ahles, T.A., Ganz, P.A., and van Dam, F.S. (2004). Cognitive impairment associated with chemotherapy for cancer: report of a workshop. J. Clin. Oncol. 22, 2233-2239.
  42. Wang, S., Yu, L., Sun, M., Mu, S., Wang, C., Wang, D., and Yao, Y. (2013). The therapeutic potential of umbilical cord mesenchymal stem cells in mice premature ovarian failure. Biomed Res. Int. 2013, 690491.
  43. Yuan, L., Wu, M.J., Sun, H.Y., Xiong, J., Zhang, Y., Liu, C.Y., Fu, L.L., Liu, D.M., Liu, H.-Q., and Mei, C.-L. (2011). VEGF-modified human embryonic mesenchymal stem cell implantation enhances protection against cisplatin-induced acute kidney injury. Am. J. Physiol. Renal. Physiol. 300, F207-F218. https://doi.org/10.1152/ajprenal.00073.2010
  44. Zhang, A., Wang, Y., Ye, Z., Xie, H., Zhou, L., and Zheng, S. (2010). Mechanism of TNF-${\alpha}$-nduced migration and hepatocyte growth factor production in human mesenchymal stem cells. J. Cell. Biochem. 111, 469-475. https://doi.org/10.1002/jcb.22729

Cited by

  1. Advances in improving fertility in women through stem cell-based clinical platforms vol.17, pp.5, 2017, https://doi.org/10.1080/14712598.2017.1305352
  2. Ginkgolide B protects against cisplatin-induced ototoxicity: enhancement of Akt–Nrf2–HO-1 signaling and reduction of NADPH oxidase vol.75, pp.5, 2015, https://doi.org/10.1007/s00280-015-2716-9
  3. miR214-regulated p53-NOX4/p66shc pathway plays a crucial role in the protective effect of Ginkgolide B against cisplatin-induced cytotoxicity in HEI-OC1 cells vol.245, 2016, https://doi.org/10.1016/j.cbi.2016.01.007
  4. Low Expression of Mfn2 Is Associated with Mitochondrial Damage and Apoptosis of Ovarian Tissues in the Premature Ovarian Failure Model vol.10, pp.9, 2015, https://doi.org/10.1371/journal.pone.0136421
  5. Mesenchymal stem cells maintain their defining stem cell characteristics after treatment with cisplatin vol.6, pp.1, 2016, https://doi.org/10.1038/srep20035
  6. VSELs may obviate cryobanking of gonadal tissue in cancer patients for fertility preservation vol.8, pp.1, 2015, https://doi.org/10.1186/s13048-015-0199-2
  7. Letter to the Editor: Rejuvenate eggs or regenerate ovary? vol.446, 2017, https://doi.org/10.1016/j.mce.2017.03.008
  8. Mesenchymal Stem Cells (MSCs) Therapy for Recovery of Fertility: a Systematic Review 2017, https://doi.org/10.1007/s12015-017-9765-x
  9. The therapeutic potential of bone marrow mesenchymal stem cells in premature ovarian failure vol.9, pp.1, 2018, https://doi.org/10.1186/s13287-018-1008-9
  10. The current understanding of mesenchymal stem cells as potential attenuators of chemotherapy-induced toxicity pp.00207136, 2018, https://doi.org/10.1002/ijc.31619
  11. Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation improves ovarian function in rats with premature ovarian insufficiency (POI) at least partly through a paracrine mechanism vol.10, pp.1, 2019, https://doi.org/10.1186/s13287-019-1136-x
  12. The Effect of Different Routes of Injection of Bone Marrow Mesenchymal Stem Cells on Parotid Glands of Rats Receiving Cisplatin: A Comparative Study vol.10, pp.2, 2014, https://doi.org/10.15283/ijsc17022
  13. Making gametes from alternate sources of stem cells: past, present and future vol.15, pp.None, 2017, https://doi.org/10.1186/s12958-017-0308-8
  14. Effects of low-intensity pulsed ultrasound (LIPUS)-pretreated human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation on primary ovarian insufficiency in rats vol.8, pp.1, 2014, https://doi.org/10.1186/s13287-017-0739-3
  15. Effect of stem cell transplantation of premature ovarian failure in animal models and patients: A meta-analysis and case report vol.15, pp.5, 2014, https://doi.org/10.3892/etm.2018.5970
  16. Treatment potential of bone marrow-derived stem cells in women with diminished ovarian reserves and premature ovarian failure vol.31, pp.3, 2014, https://doi.org/10.1097/gco.0000000000000531
  17. The Roles of Different Stem Cells in Premature Ovarian Failure vol.14, pp.None, 2014, https://doi.org/10.2174/1574888x14666190314123006
  18. Mesenchymal stem cells for restoration of ovarian function vol.46, pp.1, 2014, https://doi.org/10.5653/cerm.2019.46.1.1
  19. Distribution of the CM-Dil-Labeled Human Umbilical Cord Vein Mesenchymal Stem Cells Migrated to the Cyclophosphamide-Injured Ovaries in C57BL/6 Mice vol.23, pp.3, 2014, https://doi.org/10.29252/ibj.23.3.200
  20. The Therapeutic Potential of Mesenchymal Stromal Cells in the Treatment of Chemotherapy-Induced Tissue Damage vol.15, pp.3, 2014, https://doi.org/10.1007/s12015-019-09886-3
  21. Uterine Cells Improved Ovarian Function in a Murine Model of Ovarian Insufficiency vol.26, pp.12, 2014, https://doi.org/10.1177/1933719119875818
  22. miR-644-5p carried by bone mesenchymal stem cell-derived exosomes targets regulation of p53 to inhibit ovarian granulosa cell apoptosis vol.10, pp.1, 2014, https://doi.org/10.1186/s13287-019-1442-3
  23. Novel Approaches in Addressing Ovarian Insufficiency in 2019: Are We There Yet? vol.29, pp.None, 2014, https://doi.org/10.1177/0963689720926154
  24. Bone marrow mesenchymal stem cell-derived exosomal miR-144-5p improves rat ovarian function after chemotherapy-induced ovarian failure by targeting PTEN vol.100, pp.3, 2014, https://doi.org/10.1038/s41374-019-0321-y
  25. In Vitro Generation of Oocyte Like Cells and Their In Vivo Efficacy: How Far We have been Succeeded vol.9, pp.3, 2020, https://doi.org/10.3390/cells9030557
  26. Effectiveness of Stem Cell Therapy in the Treatment of Ovarian Disorders and Female Infertility: A Systematic Review vol.15, pp.2, 2014, https://doi.org/10.2174/1574888x14666191119122159
  27. Premature ovarian failure and tissue engineering vol.235, pp.5, 2014, https://doi.org/10.1002/jcp.29376
  28. Placenta-Derived Mesenchymal Stem Cells Restore the Ovary Function in an Ovariectomized Rat Model via an Antioxidant Effect vol.9, pp.7, 2014, https://doi.org/10.3390/antiox9070591
  29. Human embryonic stem cell-derived mesenchymal stem cells improved premature ovarian failure vol.12, pp.8, 2020, https://doi.org/10.4252/wjsc.v12.i8.857
  30. Intra-ovarian injection of platelet-rich plasma into ovarian tissue promoted rejuvenation in the rat model of premature ovarian insufficiency and restored ovulation rate via angiogenesis modulation vol.18, pp.1, 2014, https://doi.org/10.1186/s12958-020-00638-4
  31. Quantification of the CM-Dil-labeled human umbilical cord mesenchymal stem cells migrated to the dual injured uterus in SD rat vol.11, pp.1, 2014, https://doi.org/10.1186/s13287-020-01806-4
  32. hUMSCs regulate the differentiation of ovarian stromal cells via TGF-β 1 /Smad3 signaling pathway to inhibit ovarian fibrosis to repair ovarian function in POI rats vol.11, pp.1, 2014, https://doi.org/10.1186/s13287-020-01904-3
  33. Human umbilical cord mesenchymal stem cells restore the ovarian metabolome and rescue premature ovarian insufficiency in mice vol.11, pp.1, 2014, https://doi.org/10.1186/s13287-020-01972-5
  34. In Vitro Follicular Activation and Stem Cell Therapy as a Novel Treatment Strategies in Diminished Ovarian Reserve and Primary Ovarian Insufficiency vol.11, pp.None, 2014, https://doi.org/10.3389/fendo.2020.617704
  35. Stem Cell Paracrine Signaling for Treatment of Premature Ovarian Insufficiency vol.11, pp.None, 2021, https://doi.org/10.3389/fendo.2020.626322
  36. Therapeutic Potential of HUMSCs in Female Reproductive Aging vol.9, pp.None, 2021, https://doi.org/10.3389/fcell.2021.650003
  37. Mesenchymal Stem Cells in Premature Ovarian Insufficiency: Mechanisms and Prospects vol.9, pp.None, 2014, https://doi.org/10.3389/fcell.2021.718192
  38. Research Progress on the Treatment of Premature Ovarian Failure Using Mesenchymal Stem Cells: A Literature Review vol.9, pp.None, 2021, https://doi.org/10.3389/fcell.2021.749822
  39. Hot Topics on Fertility Preservation for Women and Girls-Current Research, Knowledge Gaps, and Future Possibilities vol.10, pp.8, 2014, https://doi.org/10.3390/jcm10081650
  40. Experimental study for the establishment of a chemotherapy-induced ovarian insufficiency model in rats by using cyclophosphamide combined with busulfan vol.122, pp.None, 2021, https://doi.org/10.1016/j.yrtph.2021.104915
  41. The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility vol.10, pp.9, 2014, https://doi.org/10.3390/cells10092460
  42. Effects of single and multiple transplantations of human umbilical cord mesenchymal stem cells on the recovery of ovarian function in the treatment of premature ovarian failure in mice vol.14, pp.1, 2021, https://doi.org/10.1186/s13048-021-00871-4
  43. Transplantation of human umbilical cord mesenchymal stem cells to treat premature ovarian failure vol.12, pp.1, 2021, https://doi.org/10.1186/s13287-021-02529-w
  44. Making More Womb: Clinical Perspectives Supporting the Development and Utilization of Mesenchymal Stem Cell Therapy for Endometrial Regeneration and Infertility vol.11, pp.12, 2014, https://doi.org/10.3390/jpm11121364
  45. Insights into stem cell therapy for premature ovarian insufficiency vol.5, pp.4, 2014, https://doi.org/10.4103/2096-2924.334379