Mesenchymal stem cells for restoration of ovarian function

Article information

Clin Exp Reprod Med. 2019;46(1):1-7
Publication date (electronic) : 2019 March 1
doi : https://doi.org/10.5653/cerm.2019.46.1.1
Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
Corresponding author: Sook Young Yoon Fertility Center of CHA Gangnam Medical Center, CHA University, 566 Nonhyeon-ro, Gangnam-gu, Seoul 06135, Korea Tel: +82-2-3468-2841 Fax: +82-2-3468-3466 E-mail: syyoon11@cha.ac.kr
Received 2018 December 27; Revised 2019 January 17; Accepted 2019 February 8.

Abstract

With the progress of regenerative medicine, mesenchymal stem cells (MSCs) have received attention as a way to restore ovarian function. It has been reported that MSCs derived from bone marrow, adipose, umbilical cord blood, menstrual blood, and amniotic fluid improved ovarian function. In light of previous studies and advances in this field, there are increased expectations regarding the utilization of MSCs to restore ovarian function. This review summarizes recent research into potential applications of MSCs in women with infertility or primary ovarian insufficiency, including cases where these conditions are induced by anticancer therapy.

Characteristics of mesenchymal stem cells

Mesenchymal stem cells (MSCs), which were termed more than 25 years ago [1], represent a class of cells from human [2] and mammalian bone marrow and periosteum [3] that could be isolated and expanded in culture while maintaining their in vitro capacity to be induced to form a variety of mesodermal phenotypes and tissues. The acronym MSC can be understood as referring to mesenchymal stromal cells, multipotent stromal cells, mesenchymal progenitor cells, bone marrow stromal cells, bone marrow-derived MSCs, mesenchymal precursor cells, skeletal stem cells, and multipotent mesenchymal stromal cells [4]. To promote terminological clarity, the International Society for Cellular Therapy (ISCT) has officially defined MSCs to be multipotent mesenchymal stromal cells, suggesting that this should refer to cells from stromal tissues with plastic-adherent characteristics, while reserving the term “mesenchymal stem cells” to denote the subpopulation that actually has the two cardinal properties of stem cells (self-renewal and the capacity to differentiate down multiple lineages) [5]. The ISCT proposed three criteria to define MSCs. First, MSCs must be plastic-adherent in standard culture conditions. Second, more than 95% of MSCs must express cluster of differentiation (CD)105, CD73, and CD90, while lacking expression of CD45, CD34, CD14 or CD11b, CD79a or CD19, and human leukocyte antigen (HLA) class II. Third, these cells must be able to differentiate into osteoblasts, adipocytes, and chondroblasts under standard in vitro differentiating conditions [5].

MSCs can be derived from several tissues in the adult or infant human body, including adipose tissue, peripheral blood, umbilical cord blood, banked umbilical cord blood, umbilical cord, umbilical cord membrane, umbilical cord vein, Wharton’s jelly of the umbilical cord, placenta, decidua basalis, ligamentum flavum, amniotic fluid, amniotic membrane, dental pulp, chorionic villi of the human placenta, fetal membranes, menstrual blood, breast milk, and urine [6]. MSCs display a powerful ability to regulate immune responses, including by suppressing T cell proliferation, influencing dendritic cell maturation and function, suppressing B cell proliferation and terminal differentiation, and modulating other immune cells such as natural killer cells and macrophages [7]. Another utility of MSCs in cell therapies is homing and transendothelial migration. In the circulation, MSCs are transported to the site of damage through the vascular system, by chemokine (C–C motif), adhesion molecules (P-selectin and the VCAM-1), and matrix metalloproteinases (MMPs; MMP-2 and membrane type 1 MMP) [8]. After MSCs home to damaged tissue sites for repair, they interact closely with local stimuli, such as inflammatory cytokines, ligands of Toll-like receptors, and hypoxia, which can stimulate MSCs to produce large amounts of growth factors that perform multiple functions for tissue regeneration [8]. Based on these actions of MSCs, Caplan proposed reconceptualizing the acronym MSC as referring to “medical signaling cells [9].”

General ovarian function and premature ovarian failure

Follicles are the functional units of the ovary, and consist of an oocyte and its supporting cells, such as granulosa cells, theca cells, and stromal cells. The cells in the follicle release an oocyte monthly, resulting in fertility, and produce hormones such as estradiol and progesterone to maintain women’s overall health and to sustain pregnancy [10].

Premature ovarian failure (POF; also known as primary ovarian insufficiency), which in other words represents premature menopause, is a mysterious and complicated disease. The prevalence of POF is one in 250 women under the age of 35 years and one in 100 women under the age of 40 years. The most important mechanisms in POF are follicle dysfunction and follicle depletion [11]. Although the cause of POF has not yet been fully elucidated, genetic, endocrine, paracrine, mitochondrial dysfunction–related, and metabolic factors can affect the quality of the follicular pool and oocytes [12]. Recently, POF after chemotherapy has emerged as a major long-term adverse effect of anticancer treatment, which increases the risk of infertility and degenerative health problems. Such responses to chemotherapy may be a particular problem in young women because loss of ovarian reserve is closely related to the risk of female infertility. The exact mechanism through which anticancer drugs exert ovarian toxicity has not been fully established [13], and it seems to depend on the type of drug and the type of cell tested [14]. Stroma and granulosa cells are especially strongly affected by most anticancer drugs via apoptosis. Oocytes are known to be affected by indirect toxicity through the stroma and granulosa cells that surround them. An alkylating agent, cyclophosphamide (CTX) induces double-strand breaks of DNA in actively proliferating cells, such as granulosa cells and stromal cells, and oocytes also experience damage caused by CTX and other anticancer drugs [13]. However, another interesting explanation has been proposed for the exhaustion of primordial follicles that are not proliferating and are therefore less sensitive to DNA damage. Primordial follicles in the resting state or activation were found to be controlled by the intracellular phosphatidylinositol 3-kinase (PI3K)-Akt-mTOR signaling pathway [15]. Phosphatase and tensin homolog (PTEN) is a reversible inhibitor of PI3K, and is known to be a tumor suppressor in humans [16]. Anticancer drug treatment such as CTX or cisplatin was found to induce depletion of primordial follicles via overrecruitment [17], and oocyte-specific PTEN deletion induced premature activation of the primordial follicle pool in a mouse model [18]. Therefore, the preservation of fertility and ovarian function should be considered as a major issue for reproductive women receiving chemotherapy. Several options exist for fertility preservation, such as cryopreservation of embryos, mature oocytes, or ovarian tissue. However, most protocols are for fertility preservation before anticancer therapy. Therefore, it is necessary to investigate possible ways to restore ovarian function.

MSC therapy and recovery of ovarian function

MSC therapy has been considered as a new option to treat female infertility or to restore ovarian function. Numerous studies have verified the protective effect of ovarian function resulting from administration of MSCs obtained from various cell sources in animal models of POF (Table 1) [19-48]. These studies of MSC transplantation have shown therapeutic potential through restoration of ovarian function and structure [19,49]. MSCs have been found to secrete growth factors, including vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), and hepatocyte growth factor (HGF) into culture medium [30], to reduce germ cell and stromal cell apoptosis, and to enhance folliculogenesis through improvements in the microenvironment [48]. As in other forms of cell therapy, two different administration methods have been considered for the use of MSCs to recover ovarian function. Intravenous administration through the tail vein has been introduced in mouse and rat models, and techniques for local administration into the ovaries have also been extensively developed [50]. In POF resulting from chemotherapy with an anticancer drug, transplantation of MSCs was found to induce ovarian function recovery, including estradiol production and improvements in ovarian structure [30,33]. MSCs showed a protective effect on apoptosis in stromal cell or granulosa cells in response to anticancer drugs, such as CTX or cisplatin (Table 1). Certain cytokines, including VEGF, HGF, and IGF-1 produced by MSCs may inhibit apoptosis in granulosa cells and upregulate B-cell lymphoma-2 in vivo [51]. Another expected mechanism of MSCs is their antifibrotic effects. Ovarian fibrogenesis is related to certain cytokines, including MMPs, tissue inhibitors of MMPs, transforming growth factor β-1, VEGF, and endothelin-1. MSCs may inhibit the proliferation of fibroblasts and decrease the deposition of extracellular matrix [51]. Angiogenesis is important mechanism in ovarian recovery. VEGF, fibroblast growth factor-2, and in particular angiogenin from MSCs induced neovascularization and facilitated blood perfusion of damaged ovarian tissues [44,51]. Ovarian function in POF mice recovered after human placenta-derived MSC transplantation through the regulation of regulatory T cells and associated cytokines [29]. After human amniotic MSCs were transplanted into naturally aged mice at 12–14 months, transplanted MSCs played a central role in inhibiting ovarian aging by secreting epidermal growth factor and HGF [22]. However, the mechanism through which MSC transplantation promotes ovarian function needs to be further investigated.

Studies on ovarian function supported by MSC administration

New candidate MSCs for restoring ovarian function

Multiple doses of MSCs in the range of 1–5 ×106 cells per kilogram of body weight are required for clinical application [52], and for animal experiments, quantities of 1×106 to 5×108 cells per mouse [24] or rat [30] have been used. However, obtaining MSCs from adult tissue requires appropriate donors, and in most cases invasive procedures must be performed. Furthermore, long-term culture potentially increases the risk of inducing chromosomal aberrations and heterogeneous cell populations, making it difficult to standardize protocols [52]. Human embryonic stem cells (hESCs) are derived from the inner cell mass of blastocysts, which is pluripotent and can differentiate into all three germ layers. Several studies have reported that MSCs can be derived from hESCs, and that they express MSC surface markers and differentiate into three germ layers (such as chondrocytes, osteoblasts, and adipocytes) [53]. Furthermore, it was reported that these cells exerted immunomodulatory effects in an in vitro experiment [54]. hESC-derived MSCs were found to be equivalent to bone marrow- or adipose-derived MSCs, making them an alternative source of MSCs for restoration of ovarian function [52]. However, ethical issues involving the use of human embryos remain, and concerns are still being discussed regarding the unwanted, unexpected, or uncontrolled differentiation of hESCs in transplantation. Nevertheless, clinical trials investigating the potential of therapy based on hESCs and hESC-derived MSCs for various diseases have been launched [55]. Induced pluripotent stem cells (iPSCs), could be another source of MSCs. iPSCs can be obtained with minimally invasive procedures, and avoid the ethical concerns about embryo use and hESCs. Furthermore, using autologous or HLA-matched iPSC lines may make it possible to minimize immunological problems [56,57].

Anothor candidate for MSCs is exosomes, which are membranebound biological nanoparticles secreted from cells. They circulate systemically, and carry in mRNA, long noncoding RNA, microRNA, proteins, and lipids. Stimulatory or inhibitory functional outcomes in response to exosomes have been found for processes including cell proliferation, apoptosis, cytokine production, immune modulation, and metastasis in cancer physiology [58]. Exosomes from MSC-mediated cell therapy were inroduced in numerous disease models, and have been found to promote functional recovery [59]. Exosomes derived from human adipose or umbilical cord blood MSCs improved ovarian function in a mouse model of premature ovarian insufficiency [60,61]. However, some problems with standardization in exosome isolation, characterization, and adiministration techniques still need to be resolved [59,62].

Safety issues of MSCs

The safety issues of MSCs should be addressed, because after MSC administration, mild adverse effects have been observed. The most severe adverse effect is that unfortunately, long-term cultured MSCs promote tumor growth and metastasis. A large number of cells must be produced for clinical-grade production of MSCs, requiring in vitro expansion, but MSCs at higher passages could lead to cell transformation. Depending upon the severity of disease, the optimal dose and specific administration time must be determined. It is necessary to thoroughly understand the underlying mechanisms that regulate and modulate MSCs, and appropriate administration methods should be developed.

Conclusion

MSCs have become the most efficient cell type in clinical applications of cell therapy. Multiple degenerative diseases and several immune-related diseases have been reported to respond to MSC transplantation. MSCs from several sources, including bone marrow, adipose tissue, umbilical cord, umbilical cord blood, placenta, amniotic fluid, endometrium, Wharton’s jelly, and menstrual blood, have been the subjects of successful experiments. According to many reports, MSCs could promote the recovery of ovarian function through inhibition of granulosa cell apoptosis and follicular atresia by upregulation of anti-Müllerian hormone and follicle-stimulating hormone receptor expression in granulosa cells. The ongoing research into the regenerative use of MSCs to recover ovarian function can be a source of hope to POF patients and infertile or subfertile women.

Notes

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Author contribution

Conceptualization, Data curation, Funding acquisition, Writing - original draft, review & editing: SYY

Acknowledgements

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2017R1D1A1B03028155).

References

1. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9:641–50.
2. Haynesworth SE, Goshima J, Goldberg VM, Caplan AI. Characterization of cells with osteogenic potential from human marrow. Bone 1992;13:81–8.
3. Nakahara H, Bruder SP, Goldberg VM, Caplan AI. In vivo osteochondrogenic potential of cultured cells derived from the periosteum. Clin Orthop Relat Res 1990;:223–32.
4. Samsonraj RM, Raghunath M, Nurcombe V, Hui JH, van Wijnen AJ, Cool SM. Concise review: multifaceted characterization of human mesenchymal stem cells for use in regenerative medicine. Stem Cells Transl Med 2017;6:2173–85.
5. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells: the International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–7.
6. Weiss ML, Troyer DL. Stem cells in the umbilical cord. Stem Cell Rev 2006;2:155–62.
7. Yi T, Song SU. Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications. Arch Pharm Res 2012;35:213–21.
8. Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin 2013;34:747–54.
9. Caplan AI. Mesenchymal stem cells: time to change the name! Stem Cells Transl Med 2017;6:1445–51.
10. Cosgrove CM, Salani R. Ovarian effects of radiation and cytotoxic chemotherapy damage. Best Pract Res Clin Obstet Gynaecol 2019;55:37–48.
11. Nelson LM. Clinical practice: primary ovarian insufficiency. N Engl J Med 2009;360:606–14.
12. Sukur YE, Kivancli IB, Ozmen B. Ovarian aging and premature ovarian failure. J Turk Ger Gynecol Assoc 2014;15:190–6.
13. Ben-Aharon I, Shalgi R. What lies behind chemotherapy-induced ovarian toxicity? Reproduction 2012;144:153–63.
14. Yuksel A, Bildik G, Senbabaoglu F, Akin N, Arvas M, Unal F, et al. The magnitude of gonadotoxicity of chemotherapy drugs on ovarian follicles and granulosa cells varies depending upon the category of the drugs and the type of granulosa cells. Hum Reprod 2015;30:2926–35.
15. Zheng W, Nagaraju G, Liu Z, Liu K. Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary. Mol Cell Endocrinol 2012;356:24–30.
16. Bertoldo MJ, Walters KA, Ledger WL, Gilchrist RB, Mermillod P, Locatelli Y. In-vitro regulation of primordial follicle activation: challenges for fertility preservation strategies. Reprod Biomed Online 2018;36:491–9.
17. Chang EM, Lim E, Yoon S, Jeong K, Bae S, Lee DR, et al. Cisplatin induces overactivation of the dormant primordial follicle through PTEN/AKT/FOXO3a pathway which leads to loss of ovarian reserve in mice. PLoS One 2015;10e0144245.
18. Reddy P, Liu L, Adhikari D, Jagarlamudi K, Rajareddy S, Shen Y, et al. Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science 2008;319:611–3.
19. Fazeli Z, Abedindo A, Omrani MD, Ghaderian SMH. Mesenchymal stem cells (MSCs) therapy for recovery of fertility: a systematic review. Stem Cell Rev 2018;14:1–12.
20. Gabr H, Elkheir WA, El-Gazzar A. Autologous stem cell transplantation in patients with idiopathic premature ovarian failure. J Tissue Sci Eng 2016;7(3 Suppl):27.
21. Edessy M, Hosni HN, Shady Y, Waf Y, Bakr S, Kamel M. Autologous stem cells therapy, the first baby of idiopathic premature ovarian failure. Acta Med Int 2016;3:19–23.
22. Ding C, Zou Q, Wang F, Wu H, Chen R, Lv J, et al. 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.
23. Grady ST, Watts AE, Thompson JA, Penedo MC, Konganti K, Hinrichs K. Effect of intra-ovarian injection of mesenchymal stem cells in aged mares. J Assist Reprod Genet 2018;Nov. 23. [Epub]. https://doi.org/10.1007/s10815-018-1371-6.
24. Kalhori Z, Azadbakht M, Soleimani Mehranjani M, Shariatzadeh MA. Improvement of the folliculogenesis by transplantation of bone marrow mesenchymal stromal cells in mice with induced polycystic ovary syndrome. Cytotherapy 2018;20:1445–58.
25. Feng P, Li P, Tan J. Human menstrual blood-derived stromal cells promote recovery of premature ovarian insufficiency via regulating the ECM-dependent FAK/AKT signaling. Stem Cell Rev 2018;Dec. 17. [Epub]. https://doi.org/10.1007/s12015-018-9867-0.
26. Bao R, Xu P, Wang Y, Wang J, Xiao L, Li G, et al. Bone marrow derived mesenchymal stem cells transplantation rescues premature ovarian insufficiency induced by chemotherapy. Gynecol Endocrinol 2018;34:320–6.
27. 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.
28. Mohamed SA, Shalaby SM, Abdelaziz M, Brakta S, Hill WD, Ismail N, et al. Human mesenchymal stem cells partially reverse infertility in chemotherapy-induced ovarian failure. Reprod Sci 2018;25:51–63.
29. 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;25:1073–82.
30. Elfayomy AK, Almasry SM, El-Tarhouny SA, Eldomiaty MA. Human umbilical cord blood-mesenchymal stem cells transplantation renovates the ovarian surface epithelium in a rat model of premature ovarian failure: possible direct and indirect effects. Tissue Cell 2016;48:370–82.
31. 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;79:938–47.
32. Pan Y, Zhang L, Zhang X, Hu C, Liu R. Biological and biomechanical analysis of two types of mesenchymal stem cells for intervention in chemotherapy-induced ovarian dysfunction. Arch Gynecol Obstet 2017;295:247–52.
33. Song D, Zhong Y, Qian C, Zou Q, Ou J, Shi Y, et al. Human umbilical cord mesenchymal stem cells therapy in cyclophosphamideinduced premature ovarian failure rat model. Biomed Res Int 2016;2016:2517514.
34. 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;31:1075–86.
35. 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.
36. Fouad H, Sabry D, Elsetohy K, Fathy N. Therapeutic efficacy of amniotic membrane stem cells and adipose tissue stem cells in rats with chemically induced ovarian failure. J Adv Res 2016;7:233–41.
37. 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;30:135–40.
38. 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;23:1548–57.
39. 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;37:865–72.
40. 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;9e106538.
41. Takehara Y, Yabuuchi A, Ezoe K, Kuroda T, Yamadera R, Sano C, et al. The restorative effects of adipose-derived mesenchymal stem cells on damaged ovarian function. Lab Invest 2013;93:181–93.
42. 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.
43. 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.
44. Zhang Y, Xia X, Yan J, Yan L, Lu C, Zhu X, et al. Mesenchymal stem cell-derived angiogenin promotes primodial follicle survival and angiogenesis in transplanted human ovarian tissue. Reprod Biol Endocrinol 2017;15:18.
45. Abd-Allah SH, Shalaby SM, Pasha HF, El-Shal AS, Raafat N, Shabrawy SM, et al. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy 2013;15:64–75.
46. Guo JQ, Gao X, Lin ZJ, Wu WZ, Huang LH, Dong HY, et al. BMSCs reduce rat granulosa cell apoptosis induced by cisplatin and perimenopause. BMC Cell Biol 2013;14:18.
47. 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.
48. 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;10:353–63.
49. Sheikhansari G, Aghebati-Maleki L, Nouri M, Jadidi-Niaragh F, Yousefi M. Current approaches for the treatment of premature ovarian failure with stem cell therapy. Biomed Pharmacother 2018;102:254–62.
50. De Becker A, Riet IV. Homing and migration of mesenchymal stromal cells: how to improve the efficacy of cell therapy? World J Stem Cells 2016;8:73–87.
51. He Y, Chen D, Yang L, Hou Q, Ma H, Xu X. The therapeutic potential of bone marrow mesenchymal stem cells in premature ovarian failure. Stem Cell Res Ther 2018;9:263.
52. Gadkari R, Zhao L, Teklemariam T, Hantash BM. Human embryonic stem cell derived-mesenchymal stem cells: an alternative mesenchymal stem cell source for regenerative medicine therapy. Regen Med 2014;9:453–65.
53. Lian Q, Lye E, Suan Yeo K, Khia Way Tan E, Salto-Tellez M, Liu TM, et al. Derivation of clinically compliant MSCs from CD105+, CD24- differentiated human ESCs. Stem Cells 2007;25:425–36.
54. Trivedi P, Hematti P. Derivation and immunological characterization of mesenchymal stromal cells from human embryonic stem cells. Exp Hematol 2008;36:350–9.
55. Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, et al. Ethical and safety issues of stem cell-based therapy. Int J Med Sci 2018;15:36–45.
56. de Rham C, Villard J. Potential and limitation of HLA-based banking of human pluripotent stem cells for cell therapy. J Immunol Res 2014;2014:518135.
57. Zhao C, Ikeya M. Generation and Applications of induced pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Int 2018;2018:9601623.
58. Huang-Doran I, Zhang CY, Vidal-Puig A. Extracellular vesicles: novel mediators of cell communication in metabolic disease. Trends Endocrinol Metab 2017;28:3–18.
59. Willis GR, Kourembanas S, Mitsialis SA. Toward exosome-based therapeutics: isolation, heterogeneity, and fit-for-purpose potency. Front Cardiovasc Med 2017;4:63.
60. Huang B, Lu J, Ding C, Zou Q, Wang W, Li H. Exosomes derived from human adipose mesenchymal stem cells improve ovary function of premature ovarian insufficiency by targeting SMAD. Stem Cell Res Ther 2018;9:216.
61. 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.
62. Doeppner TR, Bahr M, Hermann DM, Giebel B. Concise review: extracellular vesicles overcoming limitations of cell therapies in ischemic stroke. Stem Cells Transl Med 2017;6:2044–52.

Article information Continued

Table 1.

Studies on ovarian function supported by MSC administration

Study Cell source Subject Patient/animal Administration Journal
Gabr (2016) [20] Autologous bone marrow MSC Premature ovarian failure Patient Ovarian tissue/artery J Tissue Sci Eng
Edessy (2016) [21] Autologous bone marrow MSC Idiopathic premature ovarian failure Patient Ovarian autologous MSC injection Acta Med Int
Ding (2018) [22] Human amniotic MSC Natural aging 12–14-month-old mice Into the ovary Stem Cell Res Ther
Grady (2018) [23] Mares’ bone marrow MSC (age, 20–29 year) Natural aging Mare Intra-ovarian J Assist Reprod Genet
Kalhori (2018) [24] Mouse bone marrow Polycystic ovarian syndrome Mice Tail vein Cytotherapy
Feng (2018) [25] Human menstrual blood-derived CTX Mice Tail vein Stem Cell Rev
Bao (2018) [26] Mouse bone marrow CTX, busulfan Mice Tail intravenously Gynecol Endocrinol
Ling (2017) [27] Human amnion-derived MSC CTX Rat Tail vein Stem Cell Res Ther
Mohamed (2018) [28] Human bone marrow CTX, busulfan Mice Into the ovary Reprod Sci
Yin (2018) [29] Human placenta-derived pZP3 to produce autoimmune injury Mice Into the ovary Reprod Sci
Elfayomy (2016) [30] Human umbilical cord blood Paclitaxel Rat Direct injection into the bilateral ovary Tissue Cell
Gabr (2016) [31] Rat bone marrow CTX Rat Intravenously Microsc Res Tech
Pan (2017) [32] Human umbilical cord/human amniotic CTX Rat Into the ovary Arch Gynecol Obstet
Song (2016) [33] Human umbilical cord CTX Rat Tail intravenous/bilateral ovary Biomed Res Int
Su (2016) [34] Rat adipose-derived Tripterygium glycosides Rat Into the ovary Hum Reprod
Lai (2015) [35] Human endometrial CTX, busulfan Mice Tail vein J Transl Med
Fouad (2016) [36] Human amniotic membrane/adipose tissue CTX Rat Intravenously J Adv Res
Kilic (2014) [37] Rat bone marrow CTX Rat Intraperitoneal injection Gynecol Endocrinol
Liu (2014) [38] Human menstrual blood stem cell CTX Mice Engrafted in the ovary Stem Cells Dev
Liu (2014) [39] Rat bone marrow Cisplatin Rat Tail vein Mol Cells
Xiao (2014) [40] Amniotic fluid stem cell CTX, busulfan Mice Into the ovary PLoS One
Takehara (2013) [41] Human adipose-derived CTX Rat Into the ovary Lab Invest
Wang (2013) [42] Human umbilical cord CTX Mice Intravenously Biomed Res Int
Wang (2013) [43] Human amniotic epithelial cell CTX Mice Into the ovary Stem Cell Res Ther
Zhang (2017) [44] Human bone marrow Human ovarian tissue transplantation Immunedeficiency mice Ovarian transplantation Reprod Biol Endocrinol
Abd-Allah (2013) [45] Male rabbit bone marrow CTX Rabbit Intravenously in the earveins Cytotherapy
Guo (2013) [46] Rat bone marrow Perimenopause Rat Tail vein BMC Cell Biol
Liu (2012)[47] Human amniotic fluid CTX Mice Into the ovary Int J Med Sci
Fu (2008) [48] Rat bone marrow CTX Rat Into the bilateral ovary Cytotherapy

MSC, mesenchymal stem cell; CTX, cyclophosphamide.