Skip to main content

Human mesenchymal stem cell treatment of premature ovarian failure: new challenges and opportunities

Abstract

Premature ovarian failure (POF) is one of the common disorders found in women leading to 1% female infertility. Clinical features of POF are hypoestrogenism or estrogen deficiency, increased gonadotropin level, and, most importantly, amenorrhea. With the development of regenerative medicine, human mesenchymal stem cell (hMSC) therapy brings new prospects for POF. This study aimed to describe the types of MSCs currently available for POF therapy, their biological characteristics, and their mechanism of action. It reviewed the latest findings on POF to provide the theoretical basis for further investigation and clinical therapy.

Introduction

Premature ovarian failure (POF) is a common endocrine disease causing female infertility. It is characterized by high gonadotropin expression [follicle-stimulating hormone (FSH) ≥ 40 mIU/mL], low estradiol (E2) expression, and follicular dysplasia in women aged less than 40 years [1]. During 2007–2008, the term primary ovarian insufficiency (POI) was suggested to represent this dysfunction related to very early aging of the ovaries [2]. Causes of POF/POI are unknown and related to many complicated factors such as genetic defects [3], autoimmunity [4, 5], chemotherapy injury [6], and others; POF/POI can present as idiopathic [7]. Currently, the prevention and treatment of POF are extremely limited. The most commonly employed hormone replacement therapy cannot effectively recover ovarian function [8]. Thus, the demand for novel and effective therapeutics for POF has increased. Mesenchymal stem cells (MSCs) are highly important in regenerative medicine because of their inherent regenerative properties [9, 10]. Many reports suggested that human mesenchymal stem cell (hMSC) transplantation was a promising treatment for POF [11]. MSCs are multipotent stem cells that are easy to obtain and have poor immunogenicity [12, 13]. They can be harvested from several adult tissues, such as the bone marrow (BM), umbilical cord (UC), peripheral blood, adipose tissue (AD), placenta, and menstrual fluid [14,15,16,17,18,19,20]. They are an excellent source of growth factors/cytokines. In this study, the recently published data on hMSC treatment of POF were summarized, and precise mechanisms underlying the effect of stem cells on POF were explored.

Present situation in POF

The etiology of POF remains sophisticated, including X-chromosome abnormalities, autosomal genetic abnormalities, autoimmune disorder, and enzymatic defects. The POF induced by chemotherapy drugs is more prominent in young patients [21, 22]. The most common clinical treatment of POF is still hormone replacement therapy (HRT). HRT is indicated to reduce the risk of osteoporosis, cardiovascular diseases, and urogenital atrophy and to improve the quality of life of women with POF. However, the role of HRT in promoting fertility remains controversial. Artificial cycles can never replace natural cycles. HRT is considered unsafe in women with a history of breast cancer or ovarian cancer, and alternative measures should be employed to reduce risks and symptoms associated with POF [23, 24]. The last and the most promising measure resorted to is egg donation for most women with POF. However, egg donation resources are very scarce, and patients receiving donated eggs can never have their biological offspring. Therefore, clinicians are looking for new therapies for POF, and MSC transplantation is a promising treatment.

Treatment of POF using human MSCs

Human bone marrow MSCs

Characteristics of human bone marrow MSCs

Human bone marrow MSCs (hBMMSCs) have high proliferative potential and the ability to differentiate into adipocytes, chondrocytes, and osteoblasts. hBMMSCs were widely researched for treating tumors [25,26,27], cartilage repair [28], and myocardial infarction [29]. hBMMSCs are multipotent stem cells that can differentiate into multiple cell types, including osteoclasts, myocytes, macrophages, adipocytes, and cardiomyocytes [30]. hBMMSCs primarily derive their energy from glycolysis. The saturated fatty acid palmitate induces BMMSC apoptosis and decreases proliferation in vivo [31]. Many studies showed that hBMMSCs could regulate cytokine expression in Th1 and Th2 cells [32]. hBMMSCs or extracellular vesicle infusion decreased the expression of pro-inflammatory cytokines and chemokines [33].

Effects and mechanisms of hBMMSCs on POF (Fig. 1)

BMMSCs were the first stem cells used to evaluate the therapeutic ability of MSCs against chemotherapy-induced POF rat models [34]. BMMSC therapy was found to be protective against germ cell apoptosis and DNA damage in mice undergoing chemotherapy [35]. Cisplatin-induced granulosa cell (GC) apoptosis was reduced when BMMSCs migrated to GCs in vitro and the antral follicle count, E2 levels, and anti-Müllerian hormone (AMH) levels increased after 30-day BMMSC treatment [36, 37]. The new primordial follicles were formed, and FSH levels were near normal in mice with POF after BMMSC injection [38]. Moreover, BMMSCs can reactivate folliculogenesis [39]. BMMSCs homed in the stroma of injured ovaries and the levels of insulin-like growth factor-1 (IGF-1) and tumor necrosis factor-α (TNF-α) increased in ovaries [40]. Human angiogenin promotes primordial follicle survival and angiogenesis in transplanted human ovarian tissue [41]. The overexpression of miR-21 in BMMSCs could repair the ovarian function in rats with chemotherapy-induced POF, which was related to the inhibition of GC apoptosis by targeting phosphatase and tensin homolog deleted on chromosome ten (PTEN) and recombinant human programmed cell death 4 (PDCD4) [42]. Yang et al. demonstrated that BMMSC-derived exosomes could prevent ovarian follicular atresia in cyclophosphamide (CTX)-treated rats via the delivery of miR-144-5p. Moreover, miR-144-5p was related to the inhibition of GC apoptosis by targeting PTEN [43]. The effect of BMMSCs on POF was positive. However, the treatment efficacy was not sufficient for therapeutic purposes probably due to the loss of transplanted MSCs. Some scholars added simple BMMSC injection to other interventions and found that they had better therapeutic effects. Heat shock pretreatment is an effective method to enhance the anti-apoptotic capacity of BMMSCs and achieve a better treatment effect in POF [44]. In addition, human BMMSC-exosomes resulted in further stimulation of angiogenesis via the Akt/mTOR signaling pathway [45].

Fig. 1
figure 1

Effects and mechanisms of hBMMSCs on POF

Human placenta–derived MSCs

Characteristics of human placenta–derived MSCs

Human placenta–derived MSCs (hPMSCs) are a subpopulation of villous stromal cells isolated from the full-term placenta. They are widely present in the villous stroma and can differentiate into osteoblasts, cardiomyocytes, smooth muscle cells, adipocytes, endodermic pancreatic islet cells, liver cells, astrocytes, and ectodermic neurons [46,47,48]. hPMSCs express MSC markers such as stage-specific embryonic antigen (SSEA4), CD44, CD54, CD73, CD90, CD105, CD166, and Oct4 [49]. hPMSCs have immunomodulatory, anti-apoptotic, pro-angiogenic, and anti-fibrotic properties [50,51,52]. hPMSCs can secrete paracrine factors to exert an antioxidative effect [53] and also secrete pro-angiogenic molecules, including vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), transforming growth factor-beta (TGF-β), and IGF-1 [54]. HGF secreted by hPMSCs induced lining trophoblast migration in the placental villous microenvironment [49]. Besides, exosomes of hPMSCs also played important biological roles including enhancing angiogenesis in vitro and in vivo [55].

Effects and mechanisms of hPMSCs on POF (Fig. 2)

HPMSC transplantation is an effective method to recover ovarian function in mice with CTX-induced POF [56]. A study by Zhang et al. [57] reported reduced levels of FSH, LH, and antizona pellucida antibody (AZP Ab) in serum and increased the levels of AMH and E2 after transplantation of hPMSCs into female BALB/c mice aged 6–8 weeks. hPMSC transplantation significantly recovered the estrus cycle in the POF group and decreased GC apoptosis. The transplantation of hPMSCs suppressed GC apoptosis induced by the endoplasmic reticulum stress inositol-requiring enzyme 1α (IRE1α) signaling pathway in mice with autoimmune-induced POF [58]. In addition, a previous study suggested that hPMSC transplantation reversed ovarian function in mice with POF and decreased the ratios of Th17/Tc17 and Th17/regulatory T (Treg) cells via the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signaling pathway [59]. The PI3K/Akt signaling pathway is critical for follicular activation, oocyte quality, and GC development [60, 61]. Treg cells are vital in the regulation of autoimmunity and transplantation, producing immunomodulatory cytokines such as transforming growth factor β (TGF-β). Further, TGF-β inhibits the expression of interferon-gamma (IFN-γ). The exceeding IFN-γ levels inhibited ovulation [62]. HPMSC transplantation increased the release of anti-inflammatory cytokines and inhibiting pro-inflammatory production in mice with POF [63]. HPMSCs also inhibited T-cell responses and attenuated inflammation reactions. Epidermal growth factor (EGF) released from hPMSCs improved premature ovarian insufficiency via NRF2/HO-1 activation [64]. The derivation of germ cells, both female and male, was possible in vitro from placenta-derived MSCs [65]. To date, no studies have described the role of hPMSC-derived exosomes (hPMSCs-EXOs) in POF. However, some studies reported that hPMSCs-EXOs increased the levels of anti-inflammatory factor (IL-10) and decreased the levels of other inflammatory factors (IL-1β, IL-8, TNF-α, and NF-κB), thereby reducing the inflammatory response [66].

Fig. 2
figure 2

Effects and mechanisms of hPMSCs on POF

Human amniotic MSCs

Characteristics of human amniotic MSCs

Amnion is a waste product of perinatal tissue sources. Therefore, the procedure to obtain human amniotic MSCs (hAMSCs) is noninvasive and represents an advantageous source of cells for cell therapy. hAMSCs are derived from embryonic mesoderm. They are spindle, polygon, or star shaped; the number of long spindle-shaped cells increases significantly after subculture for 3 days [67]. More than 95% of hAMSCs expressed surface markers (CD105, CD29, CD44, CD73, CD49d, and CD90) [68]. Also, cells expressed embryonic/pluripotent stem cell markers such as Oct-4 and SSEA-4 [69]. Some studies identified that hAMSCs secreted a large number of various factors, including EGF, HGF, VEGF, IGF-1, growth differentiation factor 9 (GDF-9), bFGF, and many miRNAs [70]. Compared with hPMSCs, hAMSCs had higher expression of HGF, bFGF, and IGF-1 (>thirtyfold higher) [70]. The expansion potency of hAMSCs was higher than that of adult bone marrow–derived MSCs [17]. hAMSCs had many advantages to be used as a source of allogeneic cells for regenerative medicine, such as high rates of proliferation, self-renewal, multi-differentiation capacity, immunosuppressive and paracrine activity, anti-inflammatory effects, and anti-fibrotic properties [71,72,73,74,75,76]. hAMSCs might prevent age-related reductions in proliferation and differentiation potentials [77]. hAMSCs that expressed granulocyte chemotactic peptide 2 (GCP-2) and stromal cell–derived factor-1α (SDF-1α) via transcription activator-like effector nuclease (TALEN) enhanced angiogenesis [78]. hAMSCs influenced the mitogen-activated protein kinase signaling pathway to protect cells against oxidative stress–mediated dysfunction [79]. hAMSCs and their conditioned medium (CM) inhibited T cells, reduced the marker expression of Th1 and Th17 cell populations, promoted regulatory T cells, and reduced the cytotoxicity of natural killer cells [80, 81]. Furthermore, CM from hAMSCs had anti-inflammatory [82], anti-neoplastic [83], and immunomodulatory properties [84] and caused oxidative stress inhibition [85].

Effects and mechanisms of hAMSCs on POF (Fig. 3)

The secretion of FIGF-1, HGF, GF2, and VEGF by human AMSCs improves the ovarian function of POF. The follicle numbers were recovered to the normal level during week 4 of hAMSC transplantation in the medium-dose CTX group (70 mg/kg). hAMSCs increased ki67+AMH+ cell numbers in patients with diminished ovarian reserve (DOR) and POF (Table 1) [68]. The number of follicles and oocytes and the level of serum E2 significantly increased, but the serum FSH levels significantly decreased in mice with POF + hAMSCs. Levels of Stra8 and integrin beta-1 were upregulated compared with those in model mice [86]. H2O2 is an endogenous reactive oxygen species, and strong oxidants can cause DNA damage, inflammation, and cell and tissue injury [87,88,89]. Liu et al. established mice with POF in which bilateral ovaries were burned with 10% hydrogen peroxide. Results indicated that the ovarian function, levels of FSH and estrogen, and fertility rates were recovered in mice with POF-hAMSC transplantation, and normal newborn mice were produced [67]. In mice with natural ovarian aging, hAMSCs exerted a therapeutic effect on mouse ovarian function by increasing follicle numbers. Furthermore, hAMSCs significantly enhanced the proliferation of GCs, and the co-culture of hGCs with growth factors (EGF and HGF from hAMSCs) stimulated the proliferation rate of GCs and inhibited the apoptotic rate more effectively [90]. The efficacy of low-intensity pulsed ultrasound–pretreated hAMSCs were superior to that of normal hAMSCs [89]. Some studies have shown that the hAMSC-mediated activation of GSK3β/β-catenin signaling was dependent upon the PI3K/Akt signaling pathway [91].

Fig. 3
figure 3

Effects and mechanisms of hAMSCs on POF

Table 1 The therapeutic effects of hAMSCs on patients’ GCs with premature ovarian failure [68]

Human UC–derived MSCs

Characteristics of human UC–derived MSCs

MSCs can be isolated from various UC compartments or from the complete UC. In particular, the Wharton’s jelly–derived MSCs (WJ-MSCs) are acquired by removing the UC vessels (arteries and veins). Human UC–derived MSCs (huMSCs) have different biological characteristics, such as painless collection method and infinite self-renewal ability. huMSCs can differentiate into three different germ layers and migrate into the damaged tissue or inflamed regions, contributing to tissue repair. WJ-MSCs have higher proliferation capacity, plasticity, immunomodulatory activity, and self-renewal ability than MSCs from other origins [92]. These huMSCs are suitable candidates for allogeneic transplantation due to their high safety and abundance, shorter expansion times, and low immunogenicity. huMSCs tested positive for surface markers, including CD90, CD73, CD29, CD105, and CD44. They attenuated the inflammatory and oxidative stress, as well as reduced the expression of senescence-related proteins and microRNAs (miRs) [93]. huMSCs exhibited a similar inhibitory effect on T-cell proliferation as on hBMMSCs at a ratio of 1:10, and the percentage of migrating cells was significantly higher in huMSCs compared with BMMSCs [94]. Moreover, no correlation was found between transplanted huMSCs and the risk of tumorigenesis [95].

Effects and mechanisms of huMSCs on POF (Table 2)

In 2013, Wang et al. [12] found that umbilical MSCs (uMSCs) could treat POF in mice. The methods for transplanting huMSCs were as follows: intravenous injection (IV) and in situ ovarian microinjection (MI). Both methods of transplantation improved ovarian function, but IV was better able to restore ovarian function compared with MI [96]. Human UC vein MSCs migrated to the cyclophosphamide-injured ovaries, and 57.1%, 32.2%, and 15% were located in the medulla, cortex, and epithelium, respectively [97]. huMSCs promoted the ovarian expression of HGF, VEGF, and IGF-1 and improved ovarian reserve function [98]. The serum levels of P, E2, and IL-4 increased, but the IFN-γ, FSH, and IL-2 levels decreased following huMSC transplantation. Also, the total number of healthy follicles increased and the number of atretic follicles decreased in mice with huMSC-POF [99]. Zheng et al. [100] used cyclophosphamide to create a POF rat model, and cultured huMSCs were transplanted by tail vein injection. They found that huMSCs reduced POF caused by chemotherapy and increased nerve growth factor (NGF) and tropomyosin receptor kinase A (TrkA) levels and decreased follicle-stimulating hormone receptor (FSHR) and caspase-3 levels via the NGF/TrkA signaling pathway. Further, huMSCs improved the ovarian function after paclitaxel injection through a direct triggering effect on the ovarian epithelium and/or indirectly enriching the ovarian niche by regulating the tissue expression of TGF-β, CK 8/18, and PCNA. These molecules are essential in regulating folliculogenesis and inhibiting CASP-3-induced apoptosis [101]. With the advancement of science and technology, scientists have performed in-depth research on the huMSC treatment of POF. huMSC membranous vesicles (MVs) were detectable within the ovaries and migrated to the ovarian follicles 24 h after transplantation. HuMSC-MV transplantation might recover ovarian function by increased angiogenesis through the PI3K/Akt signaling pathway [102]. Exosomes derived from huMSCs (huMSC-EXOs) were also used to prevent and treat chemotherapy-induced GC apoptosis in vitro [103]. In 2020, Yin et al. [104] demonstrated that heme oxygenase-1 (HO-1) expressed in huMSCs was important in restoring the ovarian function, which was mediated via the activation of JNK/Bcl-2 signaling pathway–regulated autophagy and the upregulation of the circulation of CD8+CD28− T cells. Collagen scaffold loaded huMSC transplantation in mice with POF, which improved ovarian volume and number of antral follicles and promoted ovarian angiogenesis with the increased expression of CD31; the treatment effect was very significant [105]. huMSCs on a collagen scaffold (collagen/UC-MSCs) can activate primordial follicles by phosphorylation of FOXO3a and FOXO1 in vitro [106].

Table 2 Effects and mechanisms of huMSCs on POF

Human amniotic fluid MSCs (hAFMSCs)

Characteristics of hAFMSCs

Amniotic fluid (AF) is routinely obtained via trans-abdominal amniocentesis and contains fetal-derived differentiated and undifferentiated progenitor cells including MSCs. The AF can be obtained by amniotic cavity puncture, and the operation has a minimal effect on maternal and fetal tissues. Currently, hAFMSCs are used for routine prenatal genetic diagnosis [107]. The procedure to obtain hAFMSCs is noninvasive, safe, and without social controversy, consistent with human menstrual blood MSCs [108,109,110]. In vitro, they can be expanded in different media formulations and exhibit a heterogeneous morphology with obvious epithelioid and fibroblast-shaped cells [111]. hAFMSCs have a high renewal ability and can be extended for more than 250 doublings without any loss of chromosomal telomere length [112]. Furthermore, the potential of induced pluripotent stem cells (iPSCs) derived from AF was better and more effective than that of BMMSCs [113]. AFMSCs expressed pluripotency markers, including Nanog, Oct-4, and sex-determining region y-Box 2 (SOX-2) and SOX-4, and embryonic stem cell markers, including CD117, SSEA-4, TRA1–60, and TRA-1–81 [114, 115]. Some studies reported the presence of common features between primordial germ cells and AFMSCs [116]. AFMSCs are amenable for clinical application and tissue engineering due to their low immunogenicity, anti-inflammatory properties, high proliferative ability, and differentiation capacity in vitro. Also, AFMSCs lacked carcinogenesis after transplantation in nude mice.

Effects and mechanisms of hAFMSCs on POF (Fig. 4)

Some studies showed that hAFMSCs expressed multiple growth factors such as EGF, transforming growth factor beta (TGF-β), transforming growth factor alpha (TGF-α), and bone morphogenetic protein 4 (BMP-4) [117, 118]. In 2012, Liu et al. [119] proved that CD44+/CD105+ hAFMSCs transplanted into the ovaries of mice with POF survived for at least 3 weeks, and CD44+/CD105+ hAFMSCs underwent normal cycles of cell proliferation and self-renewal in ovarian tissues of mice with POF. HAFMSC-derived exosomes (hAFMSCs-EXOs) enhanced follicular regeneration, regular estrous cycles, and AMH level through the miRNA 21/PTEN/caspase 3 signaling pathway [120]. Compared with hBMMSCs, hAFMSCs secreted higher levels of exosomes. Therefore, hAFMSCs appeared to be a preferable source of exosomes for clinical applications [121]. However, hAFMSCs-EXOs has rarely been reported in studies of POF treatment, so in the future, scholars can focus on the therapeutic effects and mechanism of hAFMSCs-EXOs in POF.

Fig. 4
figure 4

Effects and mechanisms of hAFMSCs on POF

Human menstrual blood–derived MSCs

Characteristics of human menstrual blood–derived MSCs

Human menstrual blood–derived MSCs (hMB-MSCs) were discovered by Meng et al. and Cui et al., as a novel source of MSCs [19, 122]. hMB-MSCs expressed surface markers CD9, CD29, CD41a, CD44, CD59, CD73, CD90, and CD105; human telomerase reverse transcriptase (hTERT); embryonic stem cell marker OCT-4 [19]; and germ cell–specific genes, including DAZL, VASA, c-MOS (oocyte maturation factor mos), STRA8, BLIMP1 (B-lymphocyte–induced maturation protein 1), STELLA (developmental pluripotency-associated protein 3, stella-related protein), SCP3, and SCP1; GDF9, ZPA, and ZPC (zona pellucida gene family) [123]. In addition, hMB-MSCs secrete cytokine growth factors (GM-CSF and PDGF-BB), angiogenic factors (ANG-2, VEGF, HGF, and EGF), and metalloproteinases (e.g., MMP-3 and MMP-10) [19]. hMB-MSCs can differentiate into endothelial, neurocytic, cardiomyocytic, myocytic, cartilaginous, respiratory epithelial, pancreatic, adipocytic, hepatic, and osteogenic cells [124, 125]. Liu et al. demonstrated that hMB-MSCs could differentiate into ovarian tissue–like cells [126], and Lai et al. confirmed the differentiation of hMB-MSCs into germ cells [123]. hMB-MSCs were easy to access compared with hPMSCs and huMSCs. Also, they exhibited MSC-like properties. hMB-MSCs are therefore a novel source of stem cells that can be used for tissue repair [122]. hMB-MSCs also secrete the C-X-C chemokine receptor type 4 (CXCR4) and the respective receptor for stromal cell–derived factor-1 (SDF-1), which plays an important role in MSC migration [127]. Throughout these years, an increasing number of studies paid attention to hMB-MSCs because they possessed higher proliferation rates and painless procedures [124].

Effects and mechanisms of hMB-MSCs on POF (Fig. 5)

In 2014, Liu et al. [126] injected (DiO)-labeled hMB-MSCs with green fluorescence into ovaries of POF model mice and found that hMB-MSCs could survive in POF mouse ovaries for at least 14 days. The levels of AMH, FSHR, inhibin α/β, and Ki67 increased following hMB-MSC transplantation in mice with POF. Even more surprising was that the mRNA expression in mouse ovarian cells after hMB-MSC transplantation was similar to that observed in human ovarian cells, suggesting that hMB-MSCs differentiated into ovarian granulosa–like cells in ovaries of mice with POF following stimulation of the ovarian niche. hMB-MSC transplantation could reduce apoptosis of GCs and the fibrosis of the ovarian interstitium. Especially, transplanted hMB-MSCs directly migrated to the ovarian interstitium to repair ovarian function rather than directly differentiating into oocytes. Meanwhile, hMB-MSCs exerted protective effects on damaged ovaries partially by secreting FGF2 [128]. Yan et al. isolated MB-MSCs from three healthy female volunteers and co-cultured them with hGCs treated with epirubicin. They found that MB-MSCs modulated epirubicin-induced DNA damage repair to GCs by regulating protein expression [129]. hMB-MSCs combined with Bushen Tiaochong recipe improved the ovarian function of epirubicin-induced mice with POF, which might be related to inhibiting the expression of GADD45b and promoting the expression of CyclinB1 and CDC2. This therapy combined with stem cells and other treatments also provided a new research direction in POF treatment [130]. Moreover, DiI-labeled hMB-MSCs were found to be localized in the GC layer of immature follicles. hMB-MSCs improved hormone secretion in rats with POF (e.g., AMH, FSHR, FST, E2, and P4). hMB-MSC transplantation not only changed ovarian ultrastructure but also improved ovarian function [131].

Fig. 5
figure 5

Effects and mechanisms of hMB-MSCs on POF

Human adipose tissue–derived MSCs (hADMSCs)

Characteristics of human adipose tissue–derived MSCs

Adipose tissue is suggested to be an abundant and accessible source of adult MSCs. Human adipose tissue–derived MSCs (hADMSCs) were obtained from the subcutaneous adipose tissue removed during liposuction surgeries or abdominoplasties. Moreover, the high content of hADMSCs in adipose tissue excludes the need for long-term culture in vitro and reduces the risk of chromosomal abnormalities [132]. Compared with hBMMSCs, hADMSCs had higher proliferative and self-renewal abilities and exhibited higher genetic stability in long-term culture [133]. A single unique marker was not identified in hADMSCs, but hADMSCs expressed CD34, CD14, and CD45. hADMSCs inhibited T-cell proliferation by secreting a variety of soluble mediators, including prostaglandin E2 (PGE2) and IFN-γ/indoleamine 2,3-dioxygenase, and inhibiting the NF-κB pathway [134, 135]. Hypoxia-exposed exosomes derived from hADMSCs improved angiogenesis by activating the protein kinase A (PKA) signaling pathway and promoting the expression of VEGF [136].

Effects and mechanisms of hADMSCs on POF (Fig. 6)

Human adipose stem cell–derived exosomes (hADMSC-Exos) promoted the hGC proliferation rate and inhibited the hGC apoptotic rate in POI via the regulation of SMAD (SMAD2, SMAD3, and SMAD5) signaling pathway [137]. Combined treatment with hADMSCs and estrogen increased the Treg proliferation and Foxp3 and TGF-β1 mRNA expression in POI and decreased the IFN-γ mRNA expression [138]. Moreover, the combined therapy of ADMSCs, VEGF, and platelet-rich plasma improved rat ovarian function significantly more than expected in the cyclophosphamide-induced POI model. The expression of BMP4, IGF-1, and TGF-β increased in rats with POF [139]. Transplanted hADMSCs were located only in the interstitium of ovaries. hADMSCs secreted FGF2, IGF-1, HGF, and VEGF. hADMSC transplantation improved ovarian function in rats with chemotherapy-induced POI at least partly through a paracrine mechanism [140]. Obviously, the involvement of hADMSCs in the therapeutic mechanism of POF has been less explored. hADMSCs also effectively reduced fibrosis and inflammation [141]. However, whether it affects fibrosis during the treatment of POF still needs further research.

Fig. 6
figure 6

Effects and mechanisms of hADMSCs on POF

Effects of other MSCs on POF

The peritoneum mesothelial consists of the intraperitoneal or intestinal space, abdominal wall, and omentum. The peritoneum mesothelium lines body cavities and has the same origin as the ovarian surface epithelium. Besides, the repair and reconstruction of peritoneum mesothelial cells can be performed through the secretion of growth factors (bFGF and VEGF), cytokines, and extracellular matrix [142]. Characterized peritoneum MSCs (PeMSCs) were differentiated into ovarian cell–like cells using 10% human follicular fluid and 50% human cumulus-CM for 21 days, and the expression of oocyte (Zp3 and Gdf9), germ cell (Ddx4 amd Dazl), GC (Amh), and theca cell (Lhr) markers was assessed [143]. Skin-derived MSCs (SMSCs) improved the ovarian follicle microenvironment of mice with POF, and the levels of pro-inflammatory cytokines TNF-α, TGF-β, IL-8, IL-6, IL-1β, and IFN-γ significantly decreased [144].

Problems and prospects

The past years have witnessed considerable advances in the knowledge base related to the use of MSCs for regenerative medicine. Recent breakthrough discoveries in engineering MSCs have made them an ideal source for future cell therapy in POF. It is critical to ensure the safety of MSC clinical application by understanding their impacts on tumor initiation and progression. Although many experimental and clinical assays have been developed, clinical applications of MSCs have limitations, including insufficient cell sources, immunogenicity, subculture, and ethical issues. For clinical trials, the functional potential and microbiological safety of cells must be considered, and it should be ensured that cultured cells remain untransformed. Setting up a professional system to test the quality of MSC production is extremely challenging. In addition, the large variability in cell quality is derived from different donors and tissues. Thus, more reliable and effective MSCs obtained using less invasive isolation techniques have become treatment options. Hence, cell-free therapy, which uses stem cells as a source of therapeutic molecules, can be developed to treat different disease models (Fig. 7). The main effective components of MSCs should be identified in the treatment of POF, which can avoid potential side effects caused by unnecessary treatments. Exosomes, which are important messengers between cells, regulate other cells. Numerous studies have been conducted on the biological effects of exosomes secreted by stem cells [145]. On the contrary, MSC-secreted exosomes are smaller and easier to produce and have no risk of tumor formation, facilitating their comprehensive study and clinical use in the future (Fig. 7) [146, 147]. Besides, stem cells can spread rapidly to the nearby organs or tissues, and the collagen scaffolds were believed to support the survival of transplanted cells at the initial phase of transplantation in vivo. For example, transplantation of UC-MSCs on collagen scaffold activates follicles in dormant ovaries of POF patients with long history of infertility. Therefore, MSCs that can be used combined with other therapies to prompt the treatment of POF should be explored.

Fig. 7
figure 7

Human mesenchymal stem cell treatment of premature ovarian failure: new challenges and opportunities

Conclusions

The transplantation of MSCs has brought hope for patients with POF. Especially MSCs were obtained using less invasive isolation techniques because they were not only noninvasively obtained but also out of the ethical debate. In addition, cell-free therapy (therapeutic molecules from MSCs) has been extensively researched to overcome such challenges. It is expected that POF can be successfully treated using the main effective components of MSCs and combined therapy soon.

Availability of data and materials

Not applicable.

Abbreviations

AD:

Adipose tissue

AMH:

Anti-Müllerian hormone

BM:

Bone marrow

BMP-4:

Bone morphogenetic protein 4

BMP-15:

Bone morphogenetic protein 15

bFGF:

Basic fibroblast growth factor

DOR:

Diminished ovarian reserve

E2:

Estradiol

ER:

Endoplasmic reticulum

EGF:

Epidermal growth factor

FSH:

Follicle-stimulating hormone

FSHR:

Follicle-stimulating hormone receptor

GnRH:

Gonadotropin-releasing hormone

GDF-9:

Growth differentiation factor 9

GCs:

Granulosa cells

GCP-2:

Granulocyte chemotactic peptide 2

HO-1:

Heme oxygenase-1

HRT:

Hormone replacement therapy

hMSCs:

Human mesenchymal stem cells

hPMSCs:

Human placenta–derived mesenchymal stem cells

hAMSCs:

Human amniotic mesenchymal stem cells

hBMMSCs:

Human bone marrow mesenchymal stem cells

huMSCs:

Human umbilical cord–derived mesenchymal stem cells

hAFMSCs:

Human amniotic fluid mesenchymal stem cells

hADMSCs:

Human adipose tissue–derived mesenchymal stem cells

HGF:

Hepatocyte growth factor

IGF-1:

Insulin-like growth factor-1

hTERT:

Human telomerase reverse transcriptase

IFN-g:

Interferong

IRE1α:

Stress inositol-requiring enzyme 1α

LH:

Luteinizing hormone

MAP:

Mitogen-activated protein kinase

NGF:

Nerve growth factor

POF:

Premature ovarian failure

POI:

Premature ovarian insufficiency

PB:

Peripheral blood

PGE2:

Prostaglandin E2

PTEN:

Phosphatase and tensin homolog deleted on chromosome ten

PDCD4:

Recombinant human programmed cell death 4

PeMSCs:

Peritoneum mesenchymal stem cells

SDF-1α:

Stromal cell–derived factor-1α

SMSCs:

Skin-derived mesenchymal stem cells

sSSEA:

Tage-specific embryonic antigen

SOX-2:

Sex-determining region y-Box 2

TGF-α:

Transforming growth factor alpha

TGF-β :

Transforming growth factor beta

TNF-α:

Tumor necrosis factor-α

TALEN:

Transcription activator-like effector nuclease

TrkA:

Tropomyosin receptor kinase A

UC:

Umbilical cord

VEGF:

Vascular endothelial growth factor

References

  1. Coulam CB. Premature gonadal failure. Fertil Steril. 1982;38(6):645–55.

    Article  CAS  PubMed  Google Scholar 

  2. Pastore LM, Christianson MS, Stelling J, et al. Reproductive ovarian testing and the alphabet soup of diagnoses: DOR, POI, POF, POR, and FOR. J Assist Reprod Genet. 2018;35(1):17–23.

    Article  PubMed  Google Scholar 

  3. Orlandini C, Regini C, Vellucci FL, Petraglia F, Luisi S. Genes involved in the pathogenesis of premature ovarian insufficiency. Minerva Ginecol. 2015;67(5):421–30.

    CAS  PubMed  Google Scholar 

  4. Ebrahimi M, Asbagh FA. The role of autoimmunity in premature ovarian failure. Iran J Reprod Med. 2015;13(8):461.

    PubMed  PubMed Central  Google Scholar 

  5. Yan G, Schoenfeld D, Penney C, Hurxthal K, Taylor AE, Faustman D. Identification of premature ovarian failure patients with underlying autoimmunity. J Womens Health Gend Based Med. 2000;9(3):275–87.

    Article  CAS  PubMed  Google Scholar 

  6. Kenney LB, Laufer MR, Grant FD, Grier H, Diller L. High risk of infertility and long term gonadal damage in males treated with high dose cyclophosphamide for sarcoma during childhood. Cancer. 2001;91(3):613–21.

  7. Sassarini J, Lumsden MA, Critchley HO. Sex hormone replacement in ovarian failure—new treatment concepts. Best Pract Res Clin Endocrinol Metab. 2015;29(1):105. https://doi.org/10.1016/j.beem.2014.09.010.

    Article  CAS  PubMed  Google Scholar 

  8. 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(22):3198–204.

    Article  CAS  PubMed  Google Scholar 

  9. Mohammadi M, Jaafari M, Mirzaei H, Mirzaei H. Mesenchymal stem cell: a new horizon in cancer gene therapy. Cancer Gene Ther. 2016;23(9):285–6.

    Article  CAS  PubMed  Google Scholar 

  10. Yantao He, Dongmei Chen, Lingling Yang et al. The therapeutic potential of bone marrow mesenchymal stem cells in premature ovarian failure. He et al. Stem Cell Res Ther (2018) 9:263.

  11. Phinney DG, Prockop DJ. Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair—current views. Stem Cells. 2007;25(11):2896–902.

    Article  PubMed  Google Scholar 

  12. Wang S, Yu L, Sun M, Mu S, Wang C, Wang D, Yao Y. The therapeutic potential of umbilical cord mesenchymal stem cells in mice premature ovarian failure. Biomed Res Int. Biomed Res Int. 2013;2013:690491.

  13. Augello A, Kurth TB, De Bari C. Mesenchymal stem cells: a perspective from in vitro cultures to in vivo migration and niches. Eur Cell Mater. 2010;20(121):e33.

    Google Scholar 

  14. Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36:568–84.

    Article  CAS  PubMed  Google Scholar 

  15. Lee OK, Kuo TK, Chen WM, et al. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103:1669–75.

    Article  CAS  PubMed  Google Scholar 

  16. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. In ‘t Anker PS, Scherjon SA, Kleijburg-van der Keur C, et al. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338–45.

    Article  PubMed  Google Scholar 

  18. Korbling M, Anderlini P. Peripheral blood stem cell versus bone marrow allotransplantation: does the source of hematopoietic stem cells matter? Blood. 2001;98:2900–8.

    Article  CAS  PubMed  Google Scholar 

  19. Meng X, Ichim TE, Zhong J, et al. Endometrial regenerative cells: a novel stem cell population. J Transl Med. 2007;5:57. https://doi.org/10.1186/1479-5876-5-57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Manger K, Wildt L, Kalden JR, et al. Prevention of gonadal toxicity and preservation of gonadal function and fertility in young women with systemic lupus erythematosus treated by cyclophosphamide: the PREGO-Study. Autoimmun Rev. 2006;5:269–72.

    Article  CAS  PubMed  Google Scholar 

  21. Stearns V, Schneider B, Henry NL, et al. Breast cancer treatment and ovarian failure: risk factors and emerging genetic determinants. Nat Rev Cancer. 2006;6:886–93.

    Article  CAS  PubMed  Google Scholar 

  22. van Kasteren YM, Schoemaker J. Premature ovarian failure: a systematic review on therapeutic interventions to restore ovarian function and achieve pregnancy. Hum Reprod Update. 1999;5:483–92.

    Article  PubMed  Google Scholar 

  23. Deniz G, Antoine C, Liebens F, et al. Treatment of premature menopause in breast cancer patients. Acta Chir Belg. 2007;107(3):263–6.

    Article  CAS  PubMed  Google Scholar 

  24. Rossella E Nappi, Chiara Cassani, Margherita Rossi et al. Dealing with premature menopause in women at high-risk for hereditary genital and breast cancer. Minerva Ginecol 2016;68(5):602–612.

  25. Lu L, Chen G, Yang J, et al. Bone marrow mesenchymal stem cells suppress growth and promote the apoptosis of glioma U251 cells through downregulation of the PI3K/AKT signaling pathway. Biomed Pharmacother. 2019;112:108625.

    Article  CAS  PubMed  Google Scholar 

  26. Jiang S, Mo C, Guo S, et al. Human bone marrow mesenchymal stem cells-derived microRNA-205-containing exosomes impede the progression of prostate cancer through suppression of RHPN2. J Exp Clin Cancer Res. 2019;38(1):495.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen B, Yu J, Wang Q, et al. Human bone marrow mesenchymal stem cells promote gastric cancer growth via regulating c-Myc. Stem Cells Int. 2018;2018:9501747.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Shiraishi K, Kamei N, Takeuchi S, et al. Quality evaluation of human bone marrow mesenchymal stem cells for cartilage repair. Stem Cells Int. 2017;2017:8740294.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Mohanty S, Jain KG, Nandy SB, et al. Iron oxide labeling does not affect differentiation potential of human bone marrow mesenchymal stem cells exhibited by their differentiation into cardiac and neuronal cells. Mol Cell Biochem. 2018;448(1–2):17–26.

    Article  CAS  PubMed  Google Scholar 

  30. Neshati V, Mollazadeh S, Fazly Bazzaz BS, et al. MicroRNA-499a-5p promotes differentiation of human bone marrow-derived mesenchymal stem cells to cardiomyocytes. Appl Biochem Biotechnol. 2018;186(1):245–55.

    Article  CAS  PubMed  Google Scholar 

  31. Fillmore N, Huqi A, Jagdip S, et al. Effect of fatty acids on human bone marrow mesenchymal stem cell energy metabolism and survival. PLoS One. 2015;10(3):e0120257.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Castro-Manrreza ME, Montesinos JJ. Immunoregulation by mesenchymal stem cells: biological aspects and clinical applications. J Immunol Res. 2015;2015:394917.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Dabrowska S, Andrzejewska A, Strzemecki D, et al. Human bone marrow mesenchymal stem cell-derived extracellular vesicles attenuate neuroinflammation evoked by focal brain injury in rats. J Neuroinflammation. 2019;16(1):216.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. 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. https://doi.org/10.1080/14653240802035926.

    Article  CAS  PubMed  Google Scholar 

  35. Kilic S, Pinarli F, Ozogul C, et al. Protection from cyclophosphamide-induced ovarian damage with bone marrow-derived mesenchymal stem cells during puberty. Gynecol Endocrinol. 2014;30(2):135–40.

    Article  CAS  PubMed  Google Scholar 

  36. Liu J, Zhang H, Zhang Y, et al. Homing and restorative effects of bone marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats. Mol Cells. 2014;37(12):865–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Besikcioglu HE, Sarıbas GS, Ozogul C, et al. Determination of the effects of bone marrow derived mesenchymal stem cells and ovarian stromal stem cells on follicular maturation in cyclophosphamide induced ovarian failure in rats. Taiwan J Obstet Gynecol. 2019;58(1):53–9.

    Article  PubMed  Google Scholar 

  38. Badawy A, Sobh MA, Ahdy M, et al. Bone marrow mesenchymal stem cell repair of cyclophosphamide-induced ovarian insufficiency in a mouse model. Int J Womens Health. 2017;9:441–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mohamed SA, Shalaby SM, Abdelaziz M, et al. Human mesenchymal stem cells partially reverse infertility in chemotherapy-induced ovarian failure. Reprod Sci. 2018;25(1):51–63.

    Article  CAS  PubMed  Google Scholar 

  40. Gabr H, Rateb MA, El Sissy MH, et al. The effect of bone marrow-derived mesenchymal stem cells on chemotherapy induced ovarian failure in albino rats. Microsc Res Tech. 2016;79(10):938–47.

    Article  CAS  PubMed  Google Scholar 

  41. Zhang Y, Xia X, Yan J, et al. Mesenchymal stem cell-derived angiogenin promotes primodial follicle survival and angiogenesis in transplanted human ovarian tissue. Reprod Biol Endocrinol. 2017;15:18.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Xiafei F, He Y, Wang 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.

    Article  CAS  Google Scholar 

  43. Yang M, Lin L, Sha C, et al. Bone marrow mesenchymal stem cell-derived exosomal miR-144-5p improves rat ovarian function after chemotherapy-induced ovarian failure by targeting PTEN. Lab Investig. 2020;100(3):342–52.

    Article  CAS  PubMed  Google Scholar 

  44. Chen X, Wang Q, Li X, et al. Heat shock pretreatment of mesenchymal stem cells for inhibiting the apoptosis of ovarian granulosa cells enhanced the repair effect on chemotherapy-induced premature ovarian failure. Stem Cell Res Ther. 2018;9:240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Liang B, Liang J-M, Ding J-N, et al. Dimethyloxaloylglycine-stimulated human bone marrow mesenchymal stem cell-derived exosomes enhance bone regeneration through angiogenesis by targeting the AKT/mTOR pathway. Stem Cell Res Ther. 2019;10(1):335.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Alviano F, Fossati V, Marchionni C, Arpinati M, Bonsi L, Franchina M, Lanzoni G, Cantoni S, Cavallini C, Bianchi F, Tazzari PL, Pasquinelli G, Foroni L, et al. Term amniotic membrane is a high throughput source for multipotent mesenchymal stem cells with the ability to differentiate into endothelial cells in vitro. BMC Dev Biol. 2007;7:11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Wolbank S, Peterbauer A, Fahrner M, Hennerbichler S, van Griensven M, Stadler G, Redl H, Gabriel C. Dose-dependent immunomodulatory effect of human stem cells from amniotic membrane: a comparison with human mesenchymal stem cells from adipose tissue. Tissue Eng. 2007;13:1173–83.

    Article  CAS  PubMed  Google Scholar 

  48. Portmann-Lanz CB, Schoeberlein A, Portmann R, Mohr S, Rollini P, Sager R, Surbek DV. Turning placenta into brain: placental mesenchymal stem cells differentiate into neurons and oligodendrocytes. Am J Obstet Gynecol. 2010;202(294):e1–e11.

    Google Scholar 

  49. Chen CP. Placental villous mesenchymal cells trigger trophoblast invasion. Cell Adhes Migr. 2014;8(2):94–7.

    Article  Google Scholar 

  50. Roy R, Brodarac A, Kukucka M, Kurtz A, Becher PM, Julke K, et al. Cardioprotection by placenta-derived stromal cells in a murine myocardial infarction model. J Surg Res. 2013;185:70–83.

    Article  CAS  PubMed  Google Scholar 

  51. Prather WR, Toren A, Meiron M, Ofir R, Tschope C, Horwitz EM. The role of placental-derived adherent stromal cell (PLX-PAD) in the treatment of critical limb ischemia. Cytotherapy. 2009;11:427–34.

    Article  CAS  PubMed  Google Scholar 

  52. Zahavi-Goldstein E, Blumenfeld M, Fuchs-Telem D, Pinzur L, Rubin S, Aberman Z, et al. Placenta-derived PLX-PAD mesenchymal-like stromal cells are efficacious in rescuing blood flow in hind limb ischemia mouse model by a dose- and site-dependent mechanism of action. Cytotherapy. 2017;19:1438–46.

    Article  PubMed  Google Scholar 

  53. Liu SH, Huang JP, Lee RK, et al. Paracrine factors from human placental multipotent mesenchymal stromal cells protect endothelium from oxidative injury via STAT3 and manganese superoxide dismutase activation. Biol Reprod. 2010;82:905–13.

    Article  CAS  PubMed  Google Scholar 

  54. Kong P, Xie X, Li F, Liu Y, Lu Y. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats. Biochem Biophys Res Commun. 2013;438(2):410–9.

    Article  CAS  PubMed  Google Scholar 

  55. Komaki M, Numata Y, Morioka C, et al. Exosomes of human placenta-derived mesenchymal stem cells stimulate angiogenesis. Stem Cell Res Ther. 2017;8(1):219.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Li J, Yu Q, Huang H, Deng W, et al. Human chorionic plate-derived mesenchymal stem cells transplantation restores ovarian function in a chemotherapy-induced mouse model of premature ovarian failure. Stem Cell Res Ther. 2018;9(1):81.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Zhang H, Luo Q, Lu X, et al. Effects of hPMSCs on granulosa cell apoptosis and AMH expression and their role in the restoration of ovary function in premature ovarian failure mice. Stem Cell Res Ther. 2018;9:20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Li H, Zhao W, Wang L, et al. Human placenta-derived mesenchymal stem cells inhibit apoptosis of granulosa cells induced by IRE1α pathway in autoimmune POF mice. Cell Biol Int. 2019;43(8):899–909.

    Article  CAS  PubMed  Google Scholar 

  59. Yin N, Wang Y, Lu X, 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(1):37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Maidarti M, Anderson RA, Telfer EE, et al. Crosstalk between PTEN/PI3K/Akt signalling and DNA damage in the oocyte: implications for primordial follicle activation, oocyte quality and ageing. Cells. 2020;9(1):E200.

    Article  PubMed  CAS  Google Scholar 

  61. Yuan J, Deng Y, Zhang Y, et al. Bmp4 inhibits goose granulosa cell apoptosis via PI3K/AKT/Caspase-9 signaling pathway. Anim Reprod Sci. 2019;200:86–95.

    Article  CAS  PubMed  Google Scholar 

  62. Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev. 2001;12(1):53–72.

    Article  CAS  PubMed  Google Scholar 

  63. Yin N, Zhao W, Luo Q, et al. 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(7):1073–82.

    Article  CAS  PubMed  Google Scholar 

  64. Ding C, Zou Q, Wu Y, et al. EGF released from human placental mesenchymal stem cells improves premature ovarian insufficiency via NRF2/HO-1 activation. Aging (Albany NY). 2020;12(3):2992–3009.

    Article  CAS  Google Scholar 

  65. Asgari HR, Akbari M, Yazdekhasti H, et al. Comparison of human amniotic, chorionic, and umbilical cord multipotent mesenchymal stem cells regarding their capacity for differentiation toward female germ cells. Cell Reprogram. 2017;19(1):44–53.

    Article  CAS  PubMed  Google Scholar 

  66. Tao H, Chen X, Cao H, et al. Mesenchymal stem cell-derived extracellular vesicles for corneal wound repair. Stem Cells Int. 2019;2019:5738510.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Liu R, Zhang X, Fan Z, et al. Human amniotic mesenchymal stem cells improve the follicular microenvironment to recover ovarian function in premature ovarian failure mice. Stem Cell Res Ther. 2019;10(1):299.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Ding C, Li H, Wang Y, 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.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Magaña-Guerrero FS, Domínguez-López A, Martínez-Aboytes P, et al. Human amniotic membrane mesenchymal stem cells inhibit neutrophil extracellular traps through TSG-6. Sci Rep. 2017;7:12426.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. 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;39:918–26. https://doi.org/10.3892/ijmm.2017.2897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ladda M, Chairat T, Pakpoom K, Sirikul M. The immunosuppressive capacity of human mesenchymal stromal cells derived from amnion and bone marrow. Biochem Biophys Rep. 2016;8:34–40.

    Google Scholar 

  72. 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;70:729–39.

    Article  PubMed  PubMed Central  Google Scholar 

  73. 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;43:1758–68.

    Article  CAS  PubMed  Google Scholar 

  74. 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;43:430–9.

    Article  CAS  PubMed  Google Scholar 

  75. 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;45:2637–46.

    Article  PubMed  PubMed Central  Google Scholar 

  76. 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.

    PubMed  PubMed Central  Google Scholar 

  77. Fairbairn NG, Randolph MA, Redmond RW. The clinical applications of human amnion in plastic surgery. J Plast Reconstr Aesthet Surg. 2014;67:662–75.

    Article  CAS  PubMed  Google Scholar 

  78. Jeong IS, Park Y, Ryu HA, et al. Dual chemotactic factors-secreting human amniotic mesenchymal stem cells via TALEN-mediated gene editing enhanced angiogenesis. Int J Cardiol. 2018;260:156–62.

    Article  PubMed  Google Scholar 

  79. Wang Y, Ma J, Du Y, Miao J, Chen N. Human amnion-derived mesenchymal stem cells protect human bone marrow mesenchymal stem cells against oxidative stress-mediated dysfunction via ERK1/2 MAPK signaling. Mol Cells. 2016;39:186–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Li J, et al. Human amnion-derived stem cells have immunosuppressive properties on NK cells and monocytes. Cell Transplant. 2015;24:2065–76.

    Article  PubMed  Google Scholar 

  81. Pianta S, et al. Amniotic membrane mesenchymal cells-derived factors skew T cell polarization toward Treg and downregulate Th1 and Th17 cells subsets. Stem Cell Rev. 2015;11:394–407.

    Article  CAS  Google Scholar 

  82. Ono M, et al. Effects of human amnion-derived mesenchymal stromal cell transplantation in rats with radiation proctitis. Cytotherapy. 2015;17:1545–59.

    Article  PubMed  Google Scholar 

  83. Rolfo A, Giuffrida D, Giuffrida MC, Todros T, Calogero AE. New perspectives for prostate cancer treatment: in vitro inhibition of LNCaP and PC3 cell proliferation by amnion-derived mesenchymal stromal cells conditioned media. Aging Male. 2014;17:94–101.

    Article  PubMed  Google Scholar 

  84. Yan K, et al. Nitric oxide-mediated immunosuppressive effect of human amniotic membrane-derived mesenchymal stem cells on the viability and migration of microglia. Brain Res. 2014;1590:1–9.

    Article  CAS  PubMed  Google Scholar 

  85. Jiao H, Shi K, Zhang W, et al. Therapeutic potential of human amniotic membrane-derived mesenchymal stem cells in APP transgenic mice. Oncol Lett. 2016;12(3):1877–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Fouad H, Sabry D, Elsetohy K, et al. Therapeutic efficacy of amniotic membrane stem cells and adipose tissue stem cells in rats with chemically induced ovarian failure. J Adv Res. 2016;7(2):233–41.

    Article  CAS  PubMed  Google Scholar 

  87. 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:E3421–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. 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;115:427–35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Labunskyy VM, Gladyshev VN. Role of reactive oxygen species-mediated signaling in aging. Antioxid Redox Signal. 2013;19:1362–72. https://doi.org/10.1089/ars.2012.4891.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ding C, Zou Q, Wang F, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Li J-Y, Ren K-K, Zhang W-J, et al. Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway. Stem Cell Res Ther. 2019;10(1):247.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Ranjbaran H, Abediankenari S, Mohammadi M, et al. Wharton’s jelly derived-mesenchymal stem cells: isolation and characterization. Acta Med Iran. 2018;56(1):28–33.

    PubMed  Google Scholar 

  93. Rodrigues CE, Capcha JMC, de Bragança AC, et al. Human umbilical cord-derived mesenchymal stromal cells protect against premature renal senescence resulting from oxidative stress in rats with acute kidney injury. Stem Cell Res Ther. 2017;8:19.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Bartolucci J, Verdugo FJ, González PL, et al. Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: a phase 1/2 randomized controlled trial (RIMECARD trial [randomized clinical trial of intravenous infusion umbilical cord mesenchymal stem cells on cardiopathy]). Circ Res. 2017;121(10):1192–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Zhao Q, Zhang L, Wei Y, et al. Systematic comparison of hUC-MSCs at various passages reveals the variations of signatures and therapeutic effect on acute graft-versus-host disease. Stem Cell Res Ther. 2019;10(1):354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhang J, Xiong J, Fang L, et al. The protective effects of human umbilical cord mesenchymal stem cells on damaged ovarian function: a comparative study. Biosci Trends. 2016;10(4):265–76.

    Article  CAS  PubMed  Google Scholar 

  97. Jalalie L, Rezaie MJ, Jalili A, et al. Distribution of the CM-Dil-labeled human umbilical cord vein mesenchymal stem cells migrated to the cyclophosphamide-injured ovaries in C57BL/6 mice. Iran Biomed J. 2019;23(3):200–8.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Li J, Mao Q, He J, et al. Human umbilical cord mesenchymal stem cells improve the reserve function of perimenopausal ovary via a paracrine mechanism. Stem Cell Res Ther. 2017;8(1):55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Lu X, Cui J, Cui L, et al. The effects of human umbilical cord-derived mesenchymal stem cell transplantation on endometrial receptivity are associated with Th1/Th2 balance change and uNK cell expression of uterine in autoimmune premature ovarian failure mice. Stem Cell Res Ther. 2019;10:214.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Zheng Q, Fu X, Jiang J, et al. Umbilical cord mesenchymal stem cell transplantation prevents chemotherapy-induced ovarian failure via the NGF/TrkA pathway in rats. Biomed Res Int. 2019;2019:6539294.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Elfayomy AK, Almasry SM, El-Tarhouny SA, et al. 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(4):370–82.

    Article  CAS  PubMed  Google Scholar 

  102. Yang Z, Du X, Wang C, et al. Therapeutic effects of human umbilical cord mesenchymal stem cell-derived microvesicles on premature ovarian insufficiency in mice. Stem Cell Res Ther. 2019;10(1):250.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Sun L, Li D, Song K, 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(1):2552.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  104. Yin N, Wu C, Qiu J, et al. Protective properties of heme oxygenase-1 expressed in umbilical cord mesenchymal stem cells help restore the ovarian function of premature ovarian failure mice through activating the JNK/Bcl-2 signal pathway-regulated autophagy and upregulating the circulating of CD8+CD28−T cells. Stem Cell Res Ther. 2020;11:49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Yang Y, Lei L, Wang S, et al. Transplantation of umbilical cord-derived mesenchymal stem cells on a collagen scaffold improves ovarian function in a premature ovarian failure model of mice. In Vitro Cell Dev Biol Anim. 2019;55(4):302–11.

    Article  CAS  PubMed  Google Scholar 

  106. Ding L, Yan G, Wang B, et al. Transplantation of UC-MSCs on collagen scaffold activates follicles in dormant ovaries of POF patients with long history of infertility. Sci China Life Sci. 2018;61(12):1554–65.

    Article  CAS  PubMed  Google Scholar 

  107. Prusa AR, Marton E, Rosner M, Bernaschek G, Hengstschlager M. Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Hum Reprod. 2003;18:1489–93.

    Article  PubMed  Google Scholar 

  108. 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.

    Article  CAS  PubMed  Google Scholar 

  109. Soncini M, Vertua E, Gibelli L, Zorzi F, Denegri M, Albertini A, et al. Isolation and characterization of mesenchymal cells from human fetal membranes. J Tissue Eng Regen Med. 2007;1:296–305.

    Article  CAS  PubMed  Google Scholar 

  110. Parolini O, Alviano F, Bagnara GP, Bilic G, Buhring HJ, Evangelista M, et al. Concise review: isolation and characterization of cells from human term placenta: outcome of the first international workshop on placenta derived stem cells. Stem Cells. 2008;26:300–11.

    Article  PubMed  Google Scholar 

  111. Cananzi M, De Coppi P. CD117+ amniotic fluid stem cells: state of the art and future perspectives. Organ. 2012;8(3):77–88.

    Google Scholar 

  112. Antonucci I, Stuppia L, Kaneko Y, et al. Amniotic fluid as a rich source of mesenchymal stromal cells for transplantation therapy. Cell Transplant. 2011;20:789–95.

    Article  PubMed  Google Scholar 

  113. Li Q, Fan Y, Sun X, Yu Y. Generation of induced pluripotent stem cells from human amniotic fluid cells by reprogramming with two factors in feeder-free conditions. J Reprod Dev. 2013;59(1):72e7.

    Article  Google Scholar 

  114. Perin L, Sedrakyan S, Da Sacco S, De Filippo R. Characterization of human amniotic fluid stem cells and their pluripotential capability. Methods Cell Biol. 2008;86:85–99.

  115. Phermthai T, Odglun Y, Julavijitphong S, Titapant V, Chuenwattana P, Vantanasiri C, Pattanapanyasat K. A novel method to derive amniotic fluid stem cells for therapeutic purposes. BMC Cell Biol. 2010;11:79.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Antonucci I, Di Pietro R, Alfonsi M, et al. Human second trimester amniotic fluid cells are able to create embryoid body-like structures in vitro and to show typical expression profiles of embryonic and primordial germ cells. Cell Transplant. 2014;23(12):1501–15.

    Article  PubMed  Google Scholar 

  117. Liu T, Xu F, Du X, Lai D, Zhao Y, Huang Q, et al. Establishment and characterization of multi-drug resistant, prostate carcinoma-initiating stem-like cells from human prostate cancer cell lines 22RV1. Mol Cell Biochem. 2010;340:265–73.

    Article  CAS  PubMed  Google Scholar 

  118. Liu T, Zou G, Gao Y, Zhao X, Wang H, Huang Q, et al. High efficiency of reprogramming CD34(+) cells derived from human amniotic fluid into induced pluripotent stem cells with Oct4. Stem Cells Dev. 2012;21:2322–32.

    Article  CAS  PubMed  Google Scholar 

  119. Liu T, Huang Y, Guo L, et al. 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(7):592–602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Thabet E, Yusuf A, Abdelmonsif DA, et al. Extracellular vesicles miRNA-21: a potential therapeutic tool in premature ovarian dysfunction. Mol Hum Reprod. 2020;13:gaaa068.

    Google Scholar 

  121. Tracy SA, Ahmed A, Tigges JC, et al. A comparison of clinically relevant sources of mesenchymal stem cell-derived exosomes: bone marrow and amniotic fluid. J Pediatr Surg. 2019;54(1):86–90.

    Article  PubMed  Google Scholar 

  122. Cui CH, Uyama T, Miyado K, et al. Menstrual blood-derived cells confer human dystrophin expression in the murine model of Duchenne muscular dystrophy via cell fusion and myogenic transdifferentiation. Mol Biol Cell. 2007;18:1586–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Lai D, Guo Y, Zhang Q, et al. Differentiation of human menstrual blood-derived endometrial mesenchymal stem cells into oocyte-like cells. Acta Biochim Biophys Sin Shanghai. 2016;48(11):998–1005.

    Article  CAS  PubMed  Google Scholar 

  124. Khoury M, Alcayaga-Miranda F, Illanes SE, et al. The promising potential of menstrual stem cells for antenatal diagnosis and cell therapy. Front Immunol. 2014;5:205.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Alcayaga-Miranda F, Cuenca J, Luz-Crawford P, et al. Characterization of menstrual stem cells: angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Res Ther. 2015;6:32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Liu T, Huang Y, Zhang 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Chen L, Jingjing Q, Xiang C. The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Res Ther. 2019;10:1.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Wang Z, Wang Y, Yang T, et al. Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice. Stem Cell Res Ther. 2017;8:11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Yan Z, Guo F, Yuan Q, et al. Endometrial mesenchymal stem cells isolated from menstrual blood repaired epirubicin-induced damage to human ovarian granulosa cells by inhibiting the expression of Gadd45b in cell cycle pathway. Stem Cell Res Ther. 2019;10:4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Guo F, Xia T, Zhang Y, et al. Menstrual blood derived mesenchymal stem cells combined with Bushen Tiaochong recipe improved chemotherapy-induced premature ovarian failure in mice by inhibiting GADD45b expression in the cell cycle pathway. Reprod Biol Endocrinol. 2019;17(1):56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Manshadi MD, Navid S, Hoshino Y, et al. The effects of human menstrual blood stem cells-derived granulosa cells on ovarian follicle formation in a rat model of premature ovarian failure. Microsc Res Tech. 2019;82(6):635–42.

    Article  CAS  PubMed  Google Scholar 

  132. Uzbas E, May ID, Parisi AM, et al. Molecular physiognomies and applications of adipose-derived stem cells. Stem Cell Rev Rep. 2015;2:298–308. https://doi.org/10.1007/s12015-014-9578-0.

    Article  CAS  Google Scholar 

  133. Neri S, Bourin P, Peyrafitte JA, et al. Human adipose stromal cells (ASC) for the regeneration of injured cartilage display genetic stability after in vitro culture expansion. PLoS One. 2013;8:e77895.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. DelaRosa O, Lombardo E, Beraza A, et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng Part A. 2009;15:2795–806.

    Article  CAS  PubMed  Google Scholar 

  135. Shi D, Liao L, Zhang B, et al. Human adipose tissue-derived mesenchymal stem cells facilitate the immunosuppressive effect of cyclosporin A on T lymphocytes through Jagged-1-mediated inhibition of NF-κB signaling. Exp Hematol. 2011;39:214–24.

    Article  CAS  PubMed  Google Scholar 

  136. Xue CL, Shen YM, Li XC, et al. Exosomes derived from hypoxia-treated human adipose mesenchymal stem cells enhance angiogenesis through the PKA signaling pathway. Stem Cells Dev. 2018;27(7):456–65.

    Article  CAS  PubMed  Google Scholar 

  137. Huang B, Lu J, Ding C, et al. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Song K, Cai H, Zhang D, et al. Effects of human adipose-derived mesenchymal stem cells combined with estrogen on regulatory T cells in patients with premature ovarian insufficiency. Int Immunopharmacol. 2018;55:257–62.

    Article  CAS  PubMed  Google Scholar 

  139. Vural B, Duruksu G, Vural F, et al. Effects of VEGF+ mesenchymal stem cells and platelet-rich plasma on inbred rat ovarian functions in cyclophosphamide-induced premature ovarian insufficiency model. Stem Cell Rev Rep. 2019;15(4):558–73.

    Article  CAS  PubMed  Google Scholar 

  140. Ling L, Feng X, Wei T, et al. 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. Stem Cell Res Ther. 2019;10(1):46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Liu J, Ren J, Lina S, et al. Human adipose tissue-derived stem cells inhibit the activity of keloid fibroblasts and fibrosis in a keloid model by paracrine signaling. Burns. 2018;44(2):370–85.

    Article  PubMed  Google Scholar 

  142. Nagy JA. Peritoneal membrane morphology and function. Kidney Int Suppl. 1996;56:2–11.

    Google Scholar 

  143. Mirzaeian L, Eftekhari-Yazdi P, Esfandiari F, et al. Induction of mouse peritoneum mesenchymal stem cells into germ cell-like cells using follicular fluid and cumulus cells-conditioned media. Stem Cells Dev. 2019;28(8):554–64.

    Article  CAS  PubMed  Google Scholar 

  144. Lai D, Wang F, Dong Z, et al. Skin-derived mesenchymal stem cells help restore function to ovaries in a premature ovarian failure mouse model. PLoS One. 2014;9(5):e98749.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Zhang B, et al. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells. 2015;33:2158–68. https://doi.org/10.1002/stem.1771.

    Article  CAS  PubMed  Google Scholar 

  146. Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med. 2017;49(6):e346.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Zhang M, Jin K, Gao L, Zhang Z, Li F, Zhou F, Zhang L. Methods and technologies for exosome isolation and characterization. Small Methods. 2018;2:1800021.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No.81660257, 81960277), First-Class Discipline Construction Founded Project of Ning Xia Medical University (NXYLXK2017A05), 2018 Advantageous Subjects Project of NingXia Medical University (XY201807), and Key Research and Development program of Ningxia Hui Autonomous Region (2019BFG02005).

Author information

Authors and Affiliations

Authors

Contributions

YXF and RH were responsible for the concept of the review. YXF and RH have the same contribution to this article. YXF was responsible for writing the first draft of the manuscript. JJ contributed to the data acquisition of the article and revising it critically for important intellectual content by RH. FS and LJL were responsible for the critical review of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Rong Hu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fu, YX., Ji, J., Shan, F. et al. Human mesenchymal stem cell treatment of premature ovarian failure: new challenges and opportunities. Stem Cell Res Ther 12, 161 (2021). https://doi.org/10.1186/s13287-021-02212-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-021-02212-0

Keywords