Next Article in Journal
Impact of ABC Transporters in Osteosarcoma and Ewing’s Sarcoma: Which Are Involved in Chemoresistance and Which Are Not?
Next Article in Special Issue
Artificial Oocyte: Development and Potential Application
Previous Article in Journal
Radiotherapy Combined with PD-1 Inhibition Increases NK Cell Cytotoxicity towards Nasopharyngeal Carcinoma Cells
Previous Article in Special Issue
Analysis of the Reversible Impact of the Chemodrug Busulfan on Mouse Testes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility

1
College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
2
College of Veterinary and Animal Sciences, Jhang, Sub-Campus of University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
3
Faculty of Veterinary Medicine, Hawassa University, Hawassa 05, Ethiopia
*
Author to whom correspondence should be addressed.
Cells 2021, 10(9), 2460; https://doi.org/10.3390/cells10092460
Submission received: 20 August 2021 / Revised: 10 September 2021 / Accepted: 14 September 2021 / Published: 17 September 2021
(This article belongs to the Collection Stem Cell Application in Infertility)

Abstract

:
Infertility is a globally recognized issue caused by different reproductive disorders. To date, various therapeutic approaches to restore fertility have been attempted including etiology-specific medication, hormonal therapies, surgical excisions, and assisted reproductive technologies. Although these approaches produce results, however, fertility restoration is not achieved in all cases. Advances in using stem cell (SC) therapy hold a great promise for treating infertile patients due to their abilities to self-renew, differentiate, and produce different paracrine factors to regenerate the damaged or injured cells and replenish the affected germ cells. Furthermore, SCs secrete extracellular vesicles (EVs) containing biologically active molecules including nucleic acids, lipids, and proteins. EVs are involved in various physiological and pathological processes and show promising non-cellular therapeutic uses to combat infertility. Several studies have indicated that SCs and/or their derived EVs transplantation plays a crucial role in the regeneration of different segments of the reproductive system, oocyte production, and initiation of sperm production. However, available evidence triggers the need to testify the efficacy of SC transplantation or EVs injection in resolving the infertility issues of the human population. In this review, we highlight the recent literature covering the issues of infertility in females and males, with a special focus on the possible treatments by stem cells or their derived EVs.

1. Introduction

Infertility is defined as an inability or failure to conceive or carry a pregnancy to term. Infertility is one of the most important issues affecting the health and social lives of individuals [1]. Both males and females equally contribute to the etiology of infertility, and its causes include age, anatomical defects, defective ovulation, male factors, infections, environmental issues, genetic disorders, and autoimmune diseases [2,3,4,5,6]. These factors may contribute to gamete production failure, reduced sperm concentration, fertilization failure, and implantation failure.
Scientists have adopted different approaches to overcome these issues and restore or enhance fertility. The choice of therapeutic approach is generally dependent on the etiology and duration of infertility, individual age, and personal preferences [6]. Available therapeutic approaches include hormonal therapy, medication for a specific etiology, surgical excision, fertility preservation, and assisted reproductive technologies (ART). Both hormonal and non-hormonal drugs may directly or indirectly induce sexual dysfunction and impair gamete production and maturation [7]. Therefore, cryopreservation of gametes is desirable if the use of reprotoxic drugs is necessary or if the effects of drugs on gametes are irreversible. Later, these cryopreserved gametes may be used through ART. Hormonal therapy is effective in certain cases of infertility such as defective ovulation and low sperm count; however, it is associated with other health concerns such as breast cancer [8,9]. Recent data indicate that ART can resolve almost 80% of infertility issues [10]. However, ART is highly invasive and is associated with health concerns such as hyperstimulation syndrome, which is caused by the use of hormones [11,12]. Furthermore, undesired multiple pregnancies, ethical concerns, and financial constraints reduce the effectiveness of ART [13].

2. Stem Cell for Therapy

Stem cells (SCs) are undifferentiated cells with the capability of self-renewal and differentiation into a wide range of cell types and are an integral part of the body’s internal repair system. SCs can divide essentially without limit, serve to replenish damaged cells [14,15], and produce highly differentiated descendants [16]. SC can be derived from adults, embryos, and cord blood sources [17]. Pluripotent SCs are categorized into embryonic and induced pluripotent stem cells that are generated by integrating programming factors into adult differentiated cells [18]. Moreover, mesenchymal SC populations are reported to be present in nearly all postnatal tissues and organs and are responsible for growth, homeostasis, and repair of aged or damaged tissue [19]; however, they exhibit a multipotent property. SCs work by secreting factors that actively regulate tissue regeneration including growth factors and cytokines [20]. Recently, scientists have begun to explore the therapeutic potential of SCs to treat different etiologies to restore fertility. Cell-based therapy is an avenue that may be utilized to take offspring from both infertile and sterile individuals [21]. Moreover, the isolation of SCs from the testis and endometrium has aided in gamete production in cases of sterility due to therapy. In a mouse model, SC transplantations have been found to be effective in restoring fertility [22,23]. In rhesus macaques, Hermann et al. reported the production of functional sperm following SC transplantation after chemically induced sterility [24]. Paradoxically, controversies and ethical concerns regarding stem cell therapy have been raised by many authorities. The efficiency of pluripotent stem cell therapy faces some drawbacks such as homing inefficiency, tumor formation, non-functionality, or immune rejection [25,26]. Furthermore, embryonic stem cells require the manipulation of gametes, early embryonic life, or induction of abortion, which interfere with some religious, political, and ethical guidelines [27]. Therefore, stem cell research requires some policies and ethical regulations to meet the challenges in their applicability for therapeutics [28].

3. Extracellular Vesicles as Alternative Therapeutics

Fortunately, SCs can secrete extracellular vesicles (EVs) that possess the same therapeutic and regenerative effects as the parent SCs [29]. EVs are nano-vesicles that contain proteins, lipids, RNAs (mRNA, miRNA, and lncRNA), and other biomolecules that play an imperative role in paracrine signaling [30,31]. EVs such as exosomes and microvesicles (MVs) are released from all living cells constantly. MVs are released from the cell membrane via direct budding, while exosomes are formed from multivesicular endosomes or multivesicular bodies [32]. Sorting of bioactive molecules such as RNA, miRNA, lipids, and metabolites is carried out through different ways of charging and loading into the EVs, as reviewed in [33]. Differences in EV size and/or surface compounds affect the capturing by the objective or acceptor cells. For instance, micropinocytosis is technically consistent with the capture of isolated exosomes and small EVs, but not large EVs or small EV aggregates [33,34]. Moreover, EVs can be internalized and targeted to the lysosome for degradation or recycled and re-released into the extracellular fluids. EVs can also transmit information to acceptor cells by local acting at the cell membrane, particularly for cell surface communication signals [35]. However, the main characteristic of EV cargo delivery is to fuse with the plasma membrane and transmit molecules into the cytoplasm [34] (Figure 1). The EVs are a snapshot of the secreting cells and contain the secretome of the cell of origin and can be used to treat different reproductive disorders due to their ability to transfer different molecules. EVs can be used for various cell-free therapeutics to overcome the drawbacks of stem cells that were presented previously [25,26].
Consequently, in this review, we discuss recent laboratory reports investigating cell-based and EV cell-free therapies used to treat infertility and restore fertility.

4. Stem Cells and Female Reproductive Issues

Infertility in females can be caused by various disorders such as Asherman syndrome (AS), adenomyosis, endometriosis, preeclampsia, premature ovarian failure (POF), polycystic ovary syndrome (PCOS), recurrent implantation failure (RIF), and tubular blockage. SC transplantation has been shown to improve ovarian function and reserves [36], initiate endometrial regeneration, restore endometrial function, and improve pregnancy outcomes [37]. In this section, we provide an insight into etiologies associated with female infertility and evidence of fertility restoration using SCs.

4.1. Premature Ovarian Failure

POF is a multifactorial disorder affecting females <40 years age and is characterized by amenorrhea, deficient ovarian steroids, increased gonadotropin levels, and infertility [38,39]. Developing follicles are usually absent; instead, ovaries have a network of connective tissue interspersed with fibroblasts. Moreover, estrogen (E2) deficiency results in the atrophy of the mucosa lining the uterus and vagina [40,41]. The exact cause of POF remains unclear; however, it is associated with chemo- or radiotherapy, smoking, metabolic disorders (classic galactosemia), viral infections (HIV and mumps), autoimmune disorders, and genetic disorders (such as fragile X syndrome, Turner’s syndrome, and inhibin alpha gene affection) [42]. No effective therapy has been reported to date. Hormonal therapy can only provide temporary relief from E2 deficiency. Alternatives such as egg donations are often intractable due to economic and ethical concerns [43].
Recently, SC transplantation has been recognized as an effective therapeutic tool for the restoration of ovarian function [44,45,46]. The idea of exploiting the therapeutic potential of SCs in mammalian females with POF is based either on the production of new oocytes from specialized germline SCs [47] or a reduction in the magnitude of cellular apoptosis [48]. SC transplantation enhances ovarian function in cases of POF through reduced apoptosis of granulosa cells with modulation of cytokine expression [49]. Similar results have been observed using SCs derived from different sources, such as adipose tissue [50,51,52], amniotic fluid [53], bone marrow [48,54], menstrual blood [55,56], umbilical cord [45,57,58,59], and skin [60]. Transplanted SCs migrate to the site of follicular damage and initiate repair [61]. Proper ovarian function is reflected by higher E2 levels and improved development of follicles, with an increased population of antral follicles, ultimately resulting in the establishment of a successful pregnancy [58]. The structural and functional integrity of ovarian tissue is restored through cellular differentiation [53,55], improved vascular remodeling [62], reduced apoptosis [48,61], and upregulated antioxidant factors [63]. For regeneration of damaged tissue, SCs actively release various factors, including fibroblast growth factor (FGF) [56], cytokeratin 8/18, proliferating cell nuclear antigen [57], transforming growth factor beta, and vascular endothelial growth factor [53]. Fouad et al. compared the therapeutic effects of human amniotic membrane-derived mesenchymal SCs (hAM-MSCs) and adipose tissue-derived MSCs (Ad-MSCs) in chemotherapy-induced ovarian insufficiency in rats [64]. Their results indicated that both cell types restored ovarian function; however, the therapeutic efficacy of hAM-MSCs was higher than that of Ad-MSCs.
Recent studies involved the use of SC-derived condition medium or EVs for treating patients with POF [65,66,67,68,69]. Cytokines present in these secretions may be responsible for angiogenesis, apoptosis, cell cycle, immuno-modulation, and recovery of ovarian function [69]. In animal models, SC-derived exosomes improved the follicular morphology, suppressed apoptosis [70,71], and restored the ovarian function [72] of POF patients. It was observed that these effects were mediated by different types of RNAs including miR-664-5p, miR-144-5p, and miR-1246. In another study, Ding et al. reported that SC-derived EVs can ameliorate POF through reduced oxidative stress (OS) [73]. EVs derived from BM-MSCs can protect the cells against the adverse effects of peroxide through the reduction of malondialdehyde and increased expression of superoxide dismutase 1 and catalase [74]. The reduction in the magnitude of OS may be due to the protective effect of EVs on mitochondria [74,75] (Table 1).

4.2. Polycystic Ovarian Syndrome

PCOS is a common reproductive problem that is associated with endocrine and metabolic disorders [76,77]. PCOS is characterized by anovulation along with a higher level of androgen and luteinizing hormone (LH), whereas follicle-stimulating hormone (FSH) either remains normal or low [78]. Patients with PCOS have menstrual dysfunction, abnormal hair growth, acne, alopecia, enhanced libido, and a higher percentage of miscarriage. Furthermore, PCOS is associated with a higher risk of coronary artery disease, diabetes (type II and gestational), endometrial carcinoma, hyperlipidemia, and stroke [79,80,81]. The exact cause of PCOS is unknown; however, it is suspected that it may be caused due to a defect in the hypothalamo-hypophyseal axis, abnormal ovarian steroidogenesis, and insulin resistance [78].
Stein and Leventhal recognized obesity as a predisposing factor responsible for PCOS [82]. Therefore, weight loss has been identified as an important treatment for PCOS patients. For hormonal disorders, oral contraceptives can be used to reduce serum androgen and LH levels resulting in a relief of acne and induction of regular menses [83]. Insulin sensitizers are used to overcome metabolic disorders that result in a reduced level of insulin. Recently, Xie et el. reported that MSCs have the potential to improve the histopathology and function of the ovaries affected with PCOS [84]. MSCs significantly reduce the gene expression of pro-inflammatory and fibrosis-related factors. Ultimately, the ovarian function was restored due to a reduction in inflammation. In another study, it was found that MSC-derived EVs can ameliorate the effects of PCOS through shuttle transfer of miR-323-3p that promoted the growth of cumulus cells and inhibited their apoptosis [85] (Table 1).

4.3. Endometrial Injuries

The endometrium is regenerated under the influence of ovarian E2 every month throughout the reproductive cycle [86,87]. The endometrium is also regenerated after parturition and endometrial resection. Zuo et al. reported the presence of an endogenous population of SCs in the uterus including endometrial epithelial SCs, endometrial MSCs, and endometrial endothelium SCs [88]. These multipotent cells expressed SC-specific surface markers [89,90] and have an imperative role in endometrial functioning. In animal models, endometrial-derived SCs can form endometrial tissues [89], and their regenerative potential is considered vital for the human endometrium during reproductive cyclicity [91].
Endometrial injuries lead to scar formation and loss of function, intrauterine adhesions, amenorrhea, and increased incidence of infertility or miscarriage [92,93]. In mice, intrauterine infusion of platelet-rich plasma (PRP) has been reported to enhance regeneration in damaged endometrium [94]. Platelets contain α-granules that secrete cytokines, chemokines, and growth factors [95]. These proteins have a paracrine effect on SCs that can initiate cellular migration, proliferation, and angiogenesis, and in turn induce cell regeneration.
MSCs have the potential to induce the repair of damaged endometrial stromal cells [96] and differentiate into endometrial epithelial cells [97]. MSCs induce the repair mechanisms through the release of factors including FGF, insulin-like growth factor-1, transforming growth factor-beta (TGFβ1), and vascular endothelial growth factor. In rats, acute endometrial injuries have been treated through transplantation of bone marrow-derived MSCs (BM-MSCs) [98]. MSCs migrated to the injured parts and secreted FGF, resulting in enhanced cellular proliferation of the endometrium and muscle layer. Moreover, MSC-derived growth factors can facilitate the regeneration of microvasculature, restore implantation, and support embryonic development. In another study, umbilical cord-derived MSCs (UC-MSCs) initiated the repair of damaged endometrium, resulting in the restoration of fertility [93]. Interestingly, miR-340 from EVs derived from bone marrow stem cells downregulated collagen 1α1, α-SMA, and TGFβ1 in rats subjected to mechanical endometrial damage [99]. Furthermore, human umbilical cord mesenchymal stem cells derived EVs alleviated injured endometrial epithelial [100] and stromal cells [101] through increasing the Bcl-2 level and downregulating Cleaved Caspase-3 level, and it activated the PTEN/AKT signaling pathway to regulate proliferation and anti-apoptosis (Table 1).

4.4. Endometrial Atrophy

Endometrial atrophy is a rare condition in which the endometrial lining is not more than 5 mm in thickness [102]. Patients with this condition usually have poor reproductive outcomes. In many instances, the etiology of endometrial atrophy is unclear; however, prolonged use of oral contraceptives and tamoxifen risks this condition.
Chang et al. reported that patients with a thin endometrium (<7 mm) showed a satisfactory increase in endometrial growth after infusion with PRP, and they were able to achieve pregnancy [103]. Another research group observed similar outcomes using frozen embryo transfer [104]. Further investigations have demonstrated that PRP infusion promotes vascularization, as evidenced by increased vascular signals visible via the Doppler system [105].
SC therapy targeting the endometrial niche fills the cellular portion of the functional layers of the uterus. Santamaria et al. reported that SC therapy is an effective tool for the recovery of patients with endometrial atrophy [106]. Another study showed higher pregnancy and parturition rates using endometrium-derived MSCs (em-MSCs) in patients with thinned endometrium and with little or no responsiveness to the treatment with E2 [107]. Endometrial regeneration by SCs occurs via cellular differentiation and immunomodulation [108].

4.5. Repeated Implantation Failure and Recurrent Miscarriage

Repeated implantation failure (RIF) occurs when high-quality embryos produced through in vitro fertilization are repeatedly unable to implant [109]. Any abnormality of the embryo, endometrium, or immune system can lead to implantation failure. Successful implantation is dependent on the quality of the embryo, the implantation ability of the recipient endometrium, the maternal immune system [110,111,112], and paternal sperm factors. Maternal factors responsible for RIF include anatomical defects of the uterus, thrombophilia, diseases of connective tissue, endometrial thickness and non-receptivity, abnormal immune response [109], endometriosis [113], and competency of cumulus cells [114]. Embryonic factors responsible for RIF include genetic abnormalities and other intrinsic factors that impair embryonic development, hatching, and implantation. Treatment strategies should be dependent on the proper diagnosis of the factors responsible for RIF.
Recurrent miscarriage is defined as three consecutive pregnancy losses 20 weeks after the last menstruation. Causes of recurrent miscarriages include anatomical abnormalities of the uterus, antiphospholipid antibody syndrome, acquired or heritable thrombophilias, chromosomal abbreviations, environmental factors, infections, uncontrolled diabetes, unrecovered hypothyroidism, and other endocrine disorders [115,116].
Mammalian peripheral blood mononuclear cells (PBMCs), such as T- and B-lymphocytes and monocytes, exert a positive effect on the endometrium and its receptivity through cytokine secretion. Moreover, PBMCs help to establish the placenta and regulate immune tolerance during placentation [117]. Different research groups have reported that intrauterine application of PBMCs either alone [118], in medium supplemented with human chorionic gonadotropin [119], or co-cultured with luteal cells [120] resulted in significantly increased pregnancy, implantation, and live birth rates. Furthermore, Jensen et al. reported that fetal immuno-rejection can be prevented through the transfer of B-lymphocytes isolated from a normal gravid murine uterus to abortion-prone animals [121]. B-lymphocytes produce IgG-like antibodies that have a high affinity for antigens but are unable to trigger host defense mechanisms, thus protecting the fetus against maternally derived antibodies at the feto-maternal interface [122].
In addition to PBMCs, platelets are also involved in the implantation of human embryos [123]. Platelets play an imperative role in embryo–maternal communication and endometrial remodeling [124]. Moreover, platelets actively participate in corpus luteum formation by regulating neovascularization and luteinization [125]. Therefore, platelets may also help to increase the birth rate. This claim is well supported by the elevated pregnancy rate achieved following premating intrauterine infusion of PRP [126]. Similar outcomes have been observed in RIP-diagnosed patients treated with intrauterine administration of PRP [127].
Recently, SC therapy has been used to decrease abortion rates and improve reproductive outcomes. In mice, MSC transplantation resulted in improved reproductive performance through enhanced expression of interleukin 10 (IL-10) and TGF-β as well as reduced expression of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) mRNA [128].

4.6. Asherman’s Syndrome

This is an acquired gynecological disorder characterized by intrauterine adhesions, fibrosis, hypomenorrhea, amenorrhea, and infertility [129,130]. AS is caused by endometrial destruction as a result of endometritis, repeated and aggressive curettages, and endometrial tuberculosis [130,131]. Intrauterine scar formation or adhesion impairs blastocyst implantation and leads to abnormal placentation, including placenta previa and accreta, and in turn this leads to recurrent miscarriage or infertility [129]. In AS, patients usually have reduced or no menstrual bleeding, pain, and infertility [132]. Hysteroscopic techniques for treatment have been developed but have inconsistent outcomes [106].
Recent research has shown the effective use of SCs in the treatment of AS. In humans, SCs from different sources including adipose tissue [133], bone marrow [106,134,135], and menstrual blood [136] have been used for treating endometrium affected by AS. Nagori et al. achieved the first successful conception in a patient with AS [135]. Intrauterine contraceptives were administered to this patient for 6 months without any improvement. However, endometrial regeneration was induced after implantation of endometrial angiogenic SCs isolated from adult autologous SCs. In 2014, another study was performed in patients with AS caused by endometrial tuberculosis [134]. Following transplantation of bone marrow-derived SCs, their menstrual flow became normal, but endometrial thickness did not increase beyond 6.7 mm, and patients failed to conceive. In a pilot cohort study in 2016, Santamaria et al. observed an immediate change in endometrial morphology after SC therapy [106]. CD133+ bone marrow-derived SC implantation resulted in enhanced endometrial thickness and neoangiogenesis. Moreover, resumption of menstruation and several pregnancies were achieved.
Recent studies have shown SC-derived EVs as a new therapeutic approach for treating animals with AS [99,137,138]. EVs worked by reducing fibrosis, promoting angiogenesis, and immuno-modulation. In rats with AS, BM-MSC-derived EVs promoted the growth of endometrial glands and decreased the magnitude of fibrosis [139]. It was suspected that endometrial repair was initiated by the EVs through inhibition of the TGFβ1/Smad2 pathway. In 2020, Saribas et al. reported that rats with damaged endometrium due to AS can be treated using exosomes [140]. It was observed that exosomes derived from endometrial MSCs caused a reduction in fibrosis and induction of cellular proliferation and vascularization in the uterus. EVs promoted angiogenesis due to the increased expression of vascular marker CD31 and VEGF receptor 1 (Table 1).

4.7. Endometriosis

Endometriosis is an E2-dependent inflammatory condition that involves the formation of endometrial stroma and glands outside the uterine cavity [142]. Endometrial tissue may be found around the ovaries, peritoneum, and uterosacral and broad ligaments. Patients with endometriosis suffer from chronic pelvic pain accompanied by menorrhagia, dysmenorrhea, dyspareunia, dyschezia, and dysuria [143]. Moreover, endometriosis is associated with a high risk of allergic disorders [144], autoimmune diseases [145], cardiovascular diseases [146], ovarian and endometrial cancers [147,148], and OS [149]. The exact mechanism underlying endometriosis remains unclear. However, different pathological conditions have been proposed as a cause of endometriosis, including retrograde menstruation [150], embryonic rest [151], coelomic metaplasia, implantation of endometrial SCs, dysregulation of the immune system, and lymphovascular spread [152,153]. Moreover, endometriosis may be due to endometrial-derived SCs [154] or too high mobilization of SCs from other sources of the body [155].
Previous therapies for endometriosis were based on treating infertility issues via hormonal therapy, removal of ectopic endometrial tissue by laparotomy, and pain management [98]. Infertility issues were treated through the induction of hypoestrogenism, amenorrhea, or endometrial atrophy [156] using nonsteroidal anti-inflammatory drugs, hormonal contraceptives, progestogens, anti-progestogens, agonists and antagonists of gonadotropin-releasing hormone (GnRH), ethyl estradiol, and aromatase inhibitors [153,157,158]. However, these hormonal therapies are inadequate, especially for patients with infertility due to suppressed ovarian function accompanied by endometrial atrophy [142].
The recent discovery of autologous em-MSCs has led to their possible use in the treatment of endometriosis. The main purpose of using SCs is to reduce inflammation after adhesiolysis. However, the use of SCs for treating endometriosis is controversial due to their possible role in the etiology of endometriosis. Gopalakrishnan et al. demonstrated the therapeutic potential of MSCs in devising a targeted anti-angiogenic therapy for patients suffering from endometriosis [159]. Endometriotic tissues have enhanced angiogenesis, so targeting angiogenesis can be helpful in curbing the progression of the disease. In the equine endometriosis model, MSC-derived EVs showed a prominent anti-inflammatory activity. EVs contain miRNAs that stimulate cell proliferation and reduce both the expression and secretion of pro-inflammatory factors [141] (Table 1). In another study, human Wharton’s jelly SC-derived conditioned medium and cell-free lysate inhibited endometriotic cells through the activation of intrinsic apoptotic pathways [160].

4.8. Adenomyosis

Adenomyosis is a condition in which the myometrial lining is invaded by endometrial tissue, resulting in the uterine enlargement that microscopically exhibits ectopic, non-neoplastic, endometrial glands and stroma surrounded by hypertrophic and hyperplastic myometrium. Endometrial tissue, including glands and stroma, is haphazardly located deep inside the myometrium, and many researchers think of adenomyosis (i.e., “internal endometriosis”) as a variant form of endometriosis [161]. Similar changes may be observed in the rectovaginal septum [162]. The etiology or developmental events that may be responsible for adenomyosis are usually unknown. The most widely accepted theory about the etiology of adenomyosis is that it may be due to down-growth and invagination of the basal layer of the endometrium into the myometrium [162,163]. Factors such as weak smooth muscles and increased intrauterine pressure, either alone or together, may also contribute to the occurrence of adenomyosis. Moreover, adenomyosis is maintained by high E2 levels and impaired immune system-associated growth regulation of ectopic endometrial tissue [162].
Adenomyosis is usually diagnosed in women aged 40–50 years, and it is occasionally diagnosed in young women who are either undergoing infertility evaluations or have symptoms of dysmenorrhea and menorrhagia. Adenomyosis can be diagnosed using ultrasonography, magnetic resonance imaging (MRI), and histological examination of biopsy samples. Adenomyosis is responsible for abnormal bleeding, pelvic pain, and infertility.
Adenomyosis is generally treated with the same drugs used to treat endometriosis [164]. Aromatase inhibitors, contraceptive pills, danazol, dimetriose, GnRH-analogues, prostaglandin antagonists, progesterone, and Chinese herbal products have all been used to control menstrual pain and menorrhagia in patients with adenomyosis [164,165]. Furthermore, adenomyosis can be treated using uterine artery embolization [166], excision of the myometrium or adenomyoma [167], myometrial electrocoagulation [168,169], and myometrial reduction [170]. Surgical excision is preferred if drugs fail to provide relief [164]. However, uncertainty in determining the exact site and extent of adenomyosis makes it difficult to accurately remove only the affected part of the uterus; therefore, hysterectomy is the most popular procedure [165]. Chen et al. isolated adenomyosis-derived MSCs (ADS-MSCs) with higher expression of cyclooxygenase-2 (COX-2) gene [171]. He suggested that COX-2 plays a vital role in the pathogenesis of adenomyosis, as it may be responsible for controlling the migration, invasiveness, and proliferation of ADS-MSCs. Therefore, COX-2 inhibition may be helpful in treating and preventing adenomyosis.

5. Stem Cells and Male Reproductive Issues

Male infertility is defined as the inability of a male to impregnate a fertile female. Male factor infertility is usually characterized by altered sperm motility, concentration, and morphology observed in samples collected 1 and 4 weeks apart [172]. Moreover, it may involve abnormal seminal volume and accessory sex gland function [173]. Male infertility may be caused by either congenital or acquired conditions, including genetic abnormalities, hormonal disturbances, immunological disorders, infection of the reproductive tract, testicular failure, systemic diseases, cancer, varicocele, altered lifestyle, and exposure to gonadotoxic factors [174]. However, in certain cases, the etiology remains unknown [175].
Among infertile males, 15–20% exhibit azoospermia, whereas 10% have a sperm count of less than 1 million/mL of semen [176]. Much effort has been focused on treating non-obstructive azoospermia, as this is the most challenging cause of male infertility and is difficult to treat [177]. Common medical treatments used for treating male infertility issues include surgical interventions, the use of hormones, and drug therapies. However, due to their limited therapeutic effect, there is a need to seek new treatment strategies. Currently, significant advances have been made in ART for treating male infertility. Semen cryopreservation along with intra-cytoplasmic sperm injection (ICSI) is the only technique used to preserve fertility. However, its low success rate [178], as well as infertility due to the absence of functional gametes, still pose a challenge for the current ART [179]. Following the discovery of SCs, scientists have proposed the use of SCs and their associated EVs as a new therapeutic approach to treat male infertility issues [180]. Recently, SCs and SC-derived vesicles have been reported to protect and restore the fertility of sperm during freeze–thaw procedures [181,182]. The identification and isolation of spermatogonial SCs (SSCs) have led to the possibility of generating gametes. Furthermore, the production of SSCs from BM-MSCs and Ad-MSCs both in vitro [183,184] and in vivo [185,186] can greatly facilitate fertility restoration in males. Therefore, SC transplantations in infertile males with normal genetic backgrounds may be considered a promising approach for fertility restoration [187]. In this section, the different causes of male infertility and the therapeutic potential of SCs to restore fertility are explained.

5.1. Hypogonadism

Male hypogonadism is a clinical syndrome caused by the low production of testosterone (T4), sperm, or both, and it can be caused by congenital or acquired disorders. Depending on etiology, hypogonadism is categorized as either primary or secondary. Primary hypogonadism is caused by testicular disease and results in low T4 concentration, impaired spermatogenesis, and high concentrations of gonadotropins. Secondary hypogonadism is caused by dysfunction of the hypothalamic–pituitary axis and results in low T4 concentration, reduced spermatogenesis, and low concentration of gonadotropins. In primary hypogonadism, spermatogenesis is impaired more than T4 production, whereas, in secondary hypogonadism, both functions are equally affected.
The treatment of hypogonadism is usually dependent on type. Primary hypogonadism does not respond to hormonal therapy because of the damaged seminiferous tubules; ART, donor sperm, and adoption should therefore be considered instead. In patients with secondary hypogonadism, fertility can be restored using GnRH or gonadotrophin therapy [188]. Furthermore, the use of human chorionic gonadotrophin or FSH in patients with hypogonadism can result in the enlargement of the testicle [189]. In mammals, FSH use can increase the population of SSCs [190,191]. The causes of hypogonadism and their treatments especially using SCs are explained as follows.

5.1.1. Varicocele

Varicocele is an acquired cause of primary hypogonadism characterized by an abnormally enlarged collection of veins in the pampiniform plexus, which resembles a bag of worms [192]. It is the most common cause of male infertility and affects almost 15% of the population [193]. Varicocele is caused by impaired venous drainage that affects the countercurrent exchange mechanism of the testicles and results in elevated scrotal temperature and abnormal sperm production [194]. It may also result in impaired drainage of gonadotoxins from the testicles and testicular hypoxia [195]. The pathophysiology of varicocele is not well-defined; however, OS is the main factor responsible for abnormal sperm parameters. Patients with varicocele may be asymptomatic or may experience dull throbbing pain in the scrotum. Moreover, elevated temperatures may cause testicular atrophy due to the loss of germ cell mass [196].
Varicocelectomy is the first-line treatment; however, antioxidants may help to control OS [192]. In rats, berberine has been effective in varicocele treatment. Berberine resulted in improved self-renewal activities of SSCs and better Leydig–Sertoli cell communication through upregulation of BCL-6b, Etv5, and GDNF in SSCs [197]. SCs including SSCs and MSCs can inhibit apoptosis and necrosis; however, the exact antioxidant pathway is not clear [198].

5.1.2. Testicular Trauma

Testicles are predisposed to traumatic injuries because of their external location. Testicular trauma should be treated as an emergency condition because exposure of the spermatic antigen to the immune system [199] may induce an auto-immune response against sperm through the production of anti-sperm antibodies (ASA) [200]. Among infertile males, 8–21% have ASA that can negatively affect sperm motility, capacitation, and acrosome [201]. Moreover, ASA can cause sperm lysis.
The current approach to treat testicular trauma involves surgical procedures including debridement and suturing to close the tunica albuginea and avoid exposure of spermatic antigen to the immune system. The auto-immune response can be prevented by closing the tunica albuginea within a maximum of 72 h post-trauma [202]. Previous reports have reported the failure of medications or orchiectomy to prevent an autoimmune response [203], and systemic use of steroids for the treatment of immunological infertility is restricted because of side effects.
Recent reports have demonstrated that MSCs have immunomodulatory effects and may control autoimmune diseases, including animal-specific sclerosis, diabetes, graft rejection, and rheumatoid arthritis [204,205,206]. In mice, BM-MSCs transfusion resulted in immunosuppression of ASA produced following traumatic rupture of the testicle [199].

5.1.3. Testicular Torsion

Testicular torsion is considered an emergency condition [207] and can result in primary hypogonadism. Testicular torsion has a sudden onset, and patients experience intractable pain due to reduced blood supply to the testicles. Testicular torsion also results in germ cell injury, depending on the ischemia and the severity of the twist in the cord. In cases of testicular torsion, ischemia is usually followed by reperfusion, which in turn results in impaired spermatogenesis and the release of toxic substances such as free radicals from the damaged tissue into circulation [208,209,210]. During reperfusion, vascular endothelial injury and microcirculation disorders result in functionally compromised testicles.
Testicular torsion can lead to permanent dysfunction of the testicles; however, reduction and fixation of the spermatic cord within 6 h can significantly reduce its likelihood. In cases with a higher degree of twist, cellular necrosis occurs within 4 h, and a cord with a 360° twist can lead to complete or severe testicular atrophy within 24 h [211]. The recommended therapeutic approach is to ameliorate ischemic conditions, promote spermatogenesis, and regulate the immune response. Based on immunomodulatory effects, differentiation potential, and paracrine support, SCs have been reported to successfully treat torsion–induced infertility. SSCs injected into mice affected with testicular torsion restored fertility due to the upregulation of pre- and post-meiotic genes, which led to the improvement of testicular structure and function [212]. In another study, allogenic MSCs injected into torsion-induced infertile rats successfully restored fertility [207]. Hsiao et al. reported that fertility restoration in torsion-induced injury following MSC injections was the result of improved sperm motility and energy [213]. It was proposed that MSCs may be involved in the regulation of Akt/GSK3 to create a balance between glycogenesis and glycolysis in sperm. Furthermore, Zhong et al. suggested that paracrine mechanisms of UC-MSCs can protect sperm against ischemic and reperfusion injuries by reducing cellular apoptosis, inflammation, and OS [214].

5.1.4. Klinefelter’s Syndrome

Klinefelter’s syndrome (KS) is a congenital and common cause of primary hypogonadism. KS is a sex chromosome aneuploidy affecting 0.1–0.2% of newborn males. KS is characterized by small, firm testicles, androgen deficiency, azoospermia [215], and elevated gonadotropin levels [216]. The hormonal profile of KS patients is normal during their childhood, but germ cells are reduced due to the deterioration of testicles around mid-puberty [217]. KS patients had a low likelihood of fertility preservation before the introduction of ART, specifically ICSI [216]. A recent report described the extraction of microscopic sperm from almost 70% of KS patients aged 14–22 years [218]. In such patients, preserving testicular tissue containing SSCs before the onset of puberty may be helpful [219].

5.2. Epididymo-Orchitis

Epididymo-orchitis is inflammation of the epididymis and testicle, caused by infectious or non-infectious agents. Orchitis usually occurs as a result of epididymitis that spreads to the surrounding tissues [220]. Inflammation of the male genital organ affects fertility through reduced germ and Sertoli cell numbers [221]. Increased intratesticular pressure as a result of inflammation damages the germinal epithelium [194]. Different risk factors may favor epididymitis such as surgery or instrumentation of the urinary tract, prostatic obstruction, stenosis of urethral valves, long sitting hours, sexual activity, bicycle or motorcycle riding, and strenuous physical activity [222,223,224]. Based on the duration of symptoms, epididymo-orchitis can be classified as acute, subacute, or chronic [225]. The acute form persists for less than 6 weeks and is characterized by painful swelling. The chronic form persists for more than 3 months and is characterized by pain without swelling. Abscess, sepsis, and infertility are possible complications of epididymitis.
Treatment of epididymitis is usually based on the etiology. Different antibiotics, including ceftriaxone, doxycycline, azithromycin, and ofloxacin, have been reported to be effective against infectious agents [226,227]. Moreover, analgesic use, the elevation of the scrotum, reduced physical activity, and cold therapy reduces inflammation. In orchitis, supportive therapy along with the application of hot or cold packs and bed rest is recommended [225]. Antimicrobials should be avoided during viral orchitis. Erpenbach et al. and Ku et al. reported that INF-α can be used to prevent atrophy of testicles and infertility in the cases of bilateral orchitis caused by mumps [228,229]. However, Yeniyol et al. found that systemic use of INF cannot completely prevent testicular atrophy [230]. Recently, the transplantation of neonatal testis-cells combined with the use of antiviral drugs was found to be effective in restoring testicular tissue of mice with herpes virus induced-orchitis [221].

5.3. Gonadotoxicity

In males, reproductive toxicity is characterized by apoptosis of germ cells, azoospermia, and testicular atrophy [231,232]. Chemo- and radiotherapy used for treating cancer have detrimental effects on the gonads [233]. Moreover, gonadal health is negatively affected by alcohol, heavy metals, insecticides, pesticides, some antibiotics, tobacco, and recreational drugs [194]. There is a risk of permanent infertility following such treatments. In such circumstances, cryopreservation of sperm and immature testicular tissue containing SSCs is regarded as the gold standard for fertility preservation [234]. Gonadal tissue transplantation should only be considered when there is a low chance of retransmission of cancer cells. Therefore, the selection of SSCs using cell sorting techniques before transplantation is preferred [235].
In recent years, the therapeutic potential of SCs in gonadotoxicity has been investigated. MSCs have been reported to restore fertility in males with infertility induced by busulfan [186], cadmium [236], cisplatin [237], cyclophosphamide [238], and doxorubicin [239]. Similar findings were obtained in busulfan-induced azoospermic hamsters using intratubular injections of Ad-MSCs [185]. SCs can induce repair by reducing apoptosis, inflammation, and OS [237], and they can restore the normal morphology of epithelial cells lining the seminiferous tubules [186]. SCs can also reduce mitochondrial apoptosis to restore fertility [236]. Cai et al. reported that BM-MSCs restore fertility in cases of busulfan-induced azoospermia by releasing proteins, including angiogenic factors, antiapoptotic factors, chemokines, cytokines, and immunoregulators [240]. Furthermore, normal spermatogenesis was restored in the individuals with testicular toxicity using a conditioned medium derived from BM-MSCs [241] and exosomes derived from urine-derived SCs [242].
Very few recent studies have reported the use of stem cell-derived EVs for treating male infertility cases including the testicular gonadotoxicity and the non-obstructive azoospermia. We concluded these studies in Table 2. For example, testicular toxicity caused by chemotherapy or radiation was alleviated through using different kinds of stem cell-derived EVs [242,243,244,245]. Moreover, bone marrow mesenchymal stem cell-derived EVs alleviated the testicular ischemia-reperfusion injury in the rat model through reduction in apoptosis [246] (Table 2). Recently, we have reviewed the current clinical trials and animal models that use stem cells, or their derived EVs, and spermatogonial stem cells for the possible treatment of gonadotoxicity and the azoospermia cases [247].

6. Conclusions

Infertility is a global issue associated with various pathophysiological conditions. In the last few decades, the development of new therapeutic approaches for restoring fertility in infertile individuals has become an area of interest. The development of ART has led to improved treatments; however, certain cases remain untreatable. Moreover, ART is focused on preserving gametes rather than treating the source of infertility. Cellular therapy using SCs has led to alternative approaches to the treatment of infertility and restoration of fertility. Recently, cell-free therapy using stem cell-derived EVs provide a safe alternative to avoid the possible problems associated with SCs therapy. Several in vitro and in vivo studies have been performed to investigate the therapeutic potential of SCs and/or their derived EVs, which have produced satisfactory results. As most of these studies were performed on animal models, the effects of SCs and EVs therapy on the human population remain largely unknown. Hence, there is a need to further investigate the mechanisms of action, evaluate the side effects, establish quality control standards for clinical use, and legislate regulatory rules to ensure the safety of these promising therapeutic tools. We summarized the interventions and the uses of SCs and EVs for treating infertility issues in Figure 2.

Author Contributions

Conceptualization, J.C. and A.Y.Q.; methodology, J.C., A.Y.Q. and T.H.; validation, J.C., A.Y.Q. and T.H.; formal analysis, A.Y.Q., M.K.R., B.M.T. and S.B.; investigation, A.Y.Q., T.H., B.M.T., S.B. and G.S.; resources, J.C. and A.Y.Q.; data curation, A.Y.Q., T.H. and X.F.; writing—original draft preparation, A.Y.Q., M.K.R., S.B. and I.M.S.; writing—review and editing, A.Y.Q., T.H., M.K.R., B.M.T., I.M.S. and J.C.; visualization, G.S., S.B. and X.F.; supervision, J.C.; project administration, J.C.; funding acquisition, J.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Science and ICT through the National Research Foundation of Korea (NRF) (grant No. 2021R1A2C2009294) and Brain Pool program (grant No. 2021H1D3A2A02040098).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data supporting this study can be made available at reasonable request from the corresponding author.

Acknowledgments

We would like to acknowledge all the members of our research team and funding agency for their financial support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fazeli, Z.; Abedindo, A.; Omrani, M.D.; Ghaderian, S.M.H. Mesenchymal stem cells (MSCs) therapy for recovery of fertility: A systematic review. Stem Cell Rev. Rep. 2018, 14, 1–12. [Google Scholar] [CrossRef]
  2. Miyamoto, T.; Tsujimura, A.; Miyagawa, Y.; Koh, E.; Namiki, M.; Sengoku, K. Male infertility and its causes in human. Adv. Urol. 2011, 2012, 384520. [Google Scholar] [CrossRef] [Green Version]
  3. Miyamoto, T.; Minase, G.; Okabe, K.; Ueda, H.; Sengoku, K. Male infertility and its genetic causes. J. Obstet. Gynaecol. Res. 2015, 41, 1501–1505. [Google Scholar] [CrossRef]
  4. Giudice, M.G.; Del Vento, F.; Wyns, C. Male fertility preservation in DSD, XXY, pre-gonadotoxic treatments–update, methods, ethical issues, current outcomes, future directions. Best Pract. Res. Clin. Endocrinol. Metab. 2019, 33, 101261. [Google Scholar] [CrossRef] [PubMed]
  5. Gidoni, Y.; Holzer, H.; Tulandi, T.; Tan, S.L. Fertility preservation in patients with non-oncological conditions. Reprod. Biomed. Online 2008, 16, 792–800. [Google Scholar] [CrossRef]
  6. Anwar, S.; Anwar, A. Infertility: A review on causes, treatment and management. Women’s Health Gynecol. 2016, 5, 2. [Google Scholar]
  7. Semet, M.; Paci, M.; Saïas-Magnan, J.; Metzler-Guillemain, C.; Boissier, R.; Lejeune, H.; Perrin, J. The impact of drugs on male fertility: A review. Andrology 2017, 5, 640–663. [Google Scholar] [CrossRef]
  8. Holmberg, L.; Iversen, O.-E.; Rudenstam, C.M.; Hammar, M.; Kumpulainen, E.; Jaskiewicz, J.; Jassem, J.; Dobaczewska, D.; Fjosne, H.E.; Peralta, O. Increased risk of recurrence after hormone replacement therapy in breast cancer survivors. J. Natl. Cancer Inst. 2008, 100, 475–482. [Google Scholar] [CrossRef] [Green Version]
  9. Vermeulen, R.; Korse, C.; Kenter, G.; Brood-van Zanten, M.; Beurden, M.v. Safety of hormone replacement therapy following risk-reducing salpingo-oophorectomy: Systematic review of literature and guidelines. Climacteric 2019, 22, 352–360. [Google Scholar] [CrossRef]
  10. Schlegel, P. Evaluation of male infertility. Minerva Ginecol. 2009, 61, 261–283. [Google Scholar]
  11. Gómez, R.; Soares, S.R.; Busso, C.; Garcia-Velasco, J.A.; Simon, C.; Pellicer, A. Physiology and pathology of ovarian hyperstimulation syndrome. Semin. Reprod. Med. 2010, 28, 448–457. [Google Scholar] [CrossRef]
  12. Prakash, A.; Mathur, R. Ovarian hyperstimulation syndrome. Obstet. Gynaecol. 2013, 15, 31–35. [Google Scholar] [CrossRef]
  13. Van Voorhis, B.J.; Ryan, G.L. Ethical obligation for restricting the number of embryos transferred to women: Combating the multiple-birth epidemic from in vitro fertilization. Semin. Reprod. Med. 2010, 28, 287–294. [Google Scholar] [CrossRef]
  14. Garcia Cruz, D.M.; Gomes, M.; Reis, R.L.; Moratal, D.; Salmerón-Sánchez, M.; Gomez Ribelles, J.L.; Mano, J.F. Differentiation of mesenchymal stem cells in chitosan scaffolds with double micro and macroporosity. J. Biomed. Mater. Res. Part A 2010, 95, 1182–1193. [Google Scholar] [CrossRef] [Green Version]
  15. Ahn, J.S.; Ryu, H.-S.; Jung, S.-E.; Shin, B.-J.; Won, J.-H.; Um, T.G.; Oh, H.; Kim, S.-H.; Ryu, B.-Y. Expression profile of spermatogenesis associated genes in male germ cells during postnatal development in mice. J. Anim. Reprod. Biotechnol. 2020, 35, 289–296. [Google Scholar] [CrossRef]
  16. Watt, F.M.; Hogan, B.L. Out of Eden: Stem cells and their niches. Science 2000, 287, 1427–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Alison, M.R.; Poulsom, R.; Forbes, S.; Wright, N.A. An introduction to stem cells. J. Pathol. 2002, 197, 419–423. [Google Scholar] [CrossRef]
  18. Takahashi, K.; Yamanaka, S. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [Green Version]
  19. Du, H.; Taylor, H.S. Stem cells and female reproduction. Reprod. Sci. 2009, 16, 126–139. [Google Scholar]
  20. Méndez-Ferrer, S.; Michurina, T.V.; Ferraro, F.; Mazloom, A.R.; MacArthur, B.D.; Lira, S.A.; Scadden, D.T.; Ma’ayan, A.; Enikolopov, G.N.; Frenette, P.S. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010, 466, 829–834. [Google Scholar] [CrossRef]
  21. Easley, C.A., IV; Simerly, C.R.; Schatten, G. Stem cell therapeutic possibilities: Future therapeutic options for male-factor and female-factor infertility? Reprod. Biomed. Online 2013, 27, 75–80. [Google Scholar] [CrossRef] [Green Version]
  22. Brinster, R.L. Male germline stem cells: From mice to men. Science 2007, 316, 404–405. [Google Scholar] [CrossRef] [Green Version]
  23. Oatley, J.M.; Brinster, R.L. Spermatogonial stem cells. Methods Enzymol. 2006, 419, 259–282. [Google Scholar]
  24. Hermann, B.P.; Sukhwani, M.; Winkler, F.; Pascarella, J.N.; Peters, K.A.; Sheng, Y.; Valli, H.; Rodriguez, M.; Ezzelarab, M.; Dargo, G. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell 2012, 11, 715–726. [Google Scholar] [CrossRef] [Green Version]
  25. Yin, L.; Liu, X.; Shi, Y.; Ocansey, D.K.W.; Hu, Y.; Li, X.; Zhang, C.; Xu, W.; Qian, H. Therapeutic Advances of Stem Cell-Derived Extracellular Vesicles in Regenerative Medicine. Cells 2020, 9, 707. [Google Scholar] [CrossRef] [Green Version]
  26. Huang, Y.-C.; Lai, L.-C. The potential roles of stem cell-derived extracellular vesicles as a therapeutic tool. Ann. Transl. Med. 2019, 7, 693. [Google Scholar] [CrossRef]
  27. Lo, B.; Parham, L. Ethical Issues in Stem Cell Research. Endocr. Rev. 2009, 30, 204–213. [Google Scholar] [CrossRef]
  28. King, N.M.P.; Perrin, J. Ethical issues in stem cell research and therapy. Stem Cell Res. Ther. 2014, 5, 85. [Google Scholar] [CrossRef] [Green Version]
  29. Zakirova, E.Y.; Aimaletdinov, A.M.; Malanyeva, A.G.; Rutland, C.S.; Rizvanov, A.A. Extracellular Vesicles: New Perspectives of Regenerative and Reproductive Veterinary Medicine. Front. Vet. Sci. 2020, 7, 931. [Google Scholar] [CrossRef]
  30. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
  31. Ahmed, K.A.; Xiang, J. Mechanisms of cellular communication through intercellular protein transfer. J. Cell. Mol. Med. 2011, 15, 1458–1473. [Google Scholar] [CrossRef]
  32. Alzahrani, F.A.; Saadeldin, I.M. Role of Exosomes in Biological Communication Systems; Springer Nature: Singapore, 2021. [Google Scholar] [CrossRef]
  33. Al-shubaily, F.A.; Al-Zahrani, M.H. Characterization and Fine Structure of Exosomes. In Role of Exosomes in Biological Communication Systems; Springer: Singapore, 2021; pp. 27–75. [Google Scholar] [CrossRef]
  34. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  35. Tkach, M.; Kowal, J.; Zucchetti, A.E.; Enserink, L.; Jouve, M.; Lankar, D.; Saitakis, M.; Martin-Jaular, L.; Théry, C. Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes. EMBO J. 2017, 36, 3012–3028. [Google Scholar] [CrossRef] [PubMed]
  36. Yoon, S.Y. Mesenchymal stem cells for restoration of ovarian function. Clin. Exp. Reprod. Med. 2019, 46, 1. [Google Scholar] [CrossRef] [Green Version]
  37. Rungsiwiwut, R.; Virutamasen, P.; Pruksananonda, K. Mesenchymal stem cells for restoring endometrial function: An infertility perspective. Reprod. Med. Biol. 2021, 20, 13–19. [Google Scholar] [CrossRef]
  38. Nelson, L.M.; Anasti, J.N.; Flak, M.R. Premature ovarian failure. In Reproductive Endocrinology, Surgery, and Technology; Adashi, E.Y., Rock, J.A., Rosenwaks, Z., Eds.; Lippincott-Raven: Philadelphia, PA, USA, 1996; pp. 1394–1410. [Google Scholar]
  39. Luisi, S.; Orlandini, C.; Regini, C.; Pizzo, A.; Vellucci, F.; Petraglia, F. Premature ovarian insufficiency: From pathogenesis to clinical management. J. Endocrinol. Investig. 2015, 38, 597–603. [Google Scholar] [CrossRef]
  40. McGuire, M.M.; Bowden, W.; Engel, N.J.; Ahn, H.W.; Kovanci, E.; Rajkovic, A. Genomic analysis using high-resolution single-nucleotide polymorphism arrays reveals novel microdeletions associated with premature ovarian failure. Fertil. Steril. 2011, 95, 1595–1600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Duncan, M.; Cummings, L.; Chada, K. Germ cell deficient (gcd) mouse as a model of premature ovarian failure. Biol. Reprod. 1993, 49, 221–227. [Google Scholar] [CrossRef] [Green Version]
  42. Jin, M.; Yu, Y.; Huang, H. An update on primary ovarian insufficiency. Sci. China Life Sci. 2012, 55, 677–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Xiao, G.-Y.; Liu, I.-H.; Cheng, C.-C.; Chang, C.-C.; Lee, Y.-H.; Cheng, W.T.-K.; Wu, S.-C. Amniotic fluid stem cells prevent follicle atresia and rescue fertility of mice with premature ovarian failure induced by chemotherapy. PLoS ONE 2014, 9, e106538. [Google Scholar] [CrossRef]
  44. Bao, R.; Xu, P.; Wang, Y.; Wang, J.; Xiao, L.; Li, G.; Zhang, C. Bone marrow derived mesenchymal stem cells transplantation rescues premature ovarian insufficiency induced by chemotherapy. Gynecol. Endocrinol. 2018, 34, 320–326. [Google Scholar] [CrossRef] [PubMed]
  45. Song, D.; Zhong, Y.; Qian, C.; Zou, Q.; Ou, J.; Shi, Y.; Gao, L.; Wang, G.; Liu, Z.; Li, H. Human umbilical cord mesenchymal stem cells therapy in cyclophosphamide-induced premature ovarian failure rat model. BioMed. Res. Int. 2016, 2016, 2517514. [Google Scholar] [CrossRef] [Green Version]
  46. Herraiz, S.; Pellicer, N.; Romeu, M.; Pellicer, A. Treatment potential of bone marrow-derived stem cells in women with diminished ovarian reserves and premature ovarian failure. Curr. Opin. Obstet. Gynecol. 2019, 31, 156–162. [Google Scholar] [CrossRef] [PubMed]
  47. Deng, W.; Lin, H. Asymmetric germ cell division and oocyte determination during Drosophila oogenesis. Int. Rev. Cytol. 2001, 203, 93–138. [Google Scholar]
  48. Liu, J.; Zhang, H.; Zhang, Y.; Li, N.; Wen, Y.; Cao, F.; Ai, H.; Xue, X. Homing and restorative effects of bone marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats. Mol. Cells 2014, 37, 865. [Google Scholar] [CrossRef] [Green Version]
  49. Ozcan Cenksoy, P.; Oktem, M.; Erdem, O.; Karakaya, C.; Cenksoy, C.; Erdem, A.; Guner, H.; Karabacak, O. A potential novel treatment strategy: Inhibition of angiogenesis and inflammation by resveratrol for regression of endometriosis in an experimental rat model. Gynecol. Endocrinol. 2015, 31, 219–224. [Google Scholar] [CrossRef]
  50. Ding, C.; Zou, Q.; Wang, F.; Wu, H.; Wang, W.; Li, H.; Huang, B. HGF and BFGF secretion by human adipose-derived stem cells improves ovarian function during natural aging via activation of the SIRT1/FOXO1 signaling pathway. Cell. Physiol. Biochem. 2018, 45, 1316–1332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Terraciano, P.; Garcez, T.; Ayres, L.; Durli, I.; Baggio, M.; Kuhl, C.P.; Laurino, C.; Passos, E.; Paz, A.H.; Cirne-Lima, E. Cell therapy for chemically induced ovarian failure in mice. Stem Cells Int. 2014, 2014, 720753. [Google Scholar] [CrossRef]
  52. Sun, M.; Wang, S.; Li, Y.; Yu, L.; Gu, F.; Wang, C.; Yao, Y. Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure. Stem Cell Res. Ther. 2013, 4, 80. [Google Scholar] [CrossRef] [Green Version]
  53. Abd-Allah, S.H.; Shalaby, S.M.; Pasha, H.F.; Amal, S.; Raafat, N.; Shabrawy, S.M.; Awad, H.A.; Amer, M.G.; Gharib, M.A.; El Gendy, E.A. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy 2013, 15, 64–75. [Google Scholar] [CrossRef] [Green Version]
  54. Johnson, J.; Bagley, J.; Skaznik-Wikiel, M.; Lee, H.-J.; Adams, G.B.; Niikura, Y.; Tschudy, K.S.; Tilly, J.C.; Cortes, M.L.; Forkert, R. Oocyte generation in adult mammalian ovaries by putative germ cells in bone marrow and peripheral blood. Cell 2005, 122, 303–315. [Google Scholar] [CrossRef] [Green Version]
  55. Liu, T.; Huang, Y.; Zhang, J.; Qin, W.; Chi, H.; Chen, J.; Yu, Z.; Chen, C. Transplantation of human menstrual blood stem cells to treat premature ovarian failure in mouse model. Stem Cells Dev. 2014, 23, 1548–1557. [Google Scholar] [CrossRef] [Green Version]
  56. Wang, Z.; Wang, Y.; Yang, T.; Li, J.; Yang, X. Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice. Stem Cell Res. Ther. 2017, 8, 11. [Google Scholar] [CrossRef] [Green Version]
  57. Elfayomy, A.K.; Almasry, S.M.; El-Tarhouny, S.A.; Eldomiaty, M.A. 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, 370–382. [Google Scholar] [CrossRef] [PubMed]
  58. Ding, L.; Yan, G.; Wang, B.; Xu, L.; Gu, Y.; Ru, T.; Cui, X.; Lei, L.; Liu, J.; Sheng, X. 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, 1554–1565. [Google Scholar] [CrossRef] [PubMed]
  59. 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. 2013, 2013, 690491. [Google Scholar] [CrossRef]
  60. Lai, D.; Wang, F.; Dong, Z.; Zhang, Q. Skin-derived mesenchymal stem cells help restore function to ovaries in a premature ovarian failure mouse model. PLoS ONE 2014, 9, e98749. [Google Scholar] [CrossRef]
  61. 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, 135–140. [Google Scholar] [CrossRef]
  62. Cho, J.; Kim, T.-H.; Seok, J.; Jun, J.H.; Park, H.; Kweon, M.; Lim, J.-Y.; Kim, G.J. Vascular remodeling by placenta-derived mesenchymal stem cells restores ovarian function in ovariectomized rat model via the VEGF pathway. Lab. Investig. 2021, 101, 304–317. [Google Scholar] [CrossRef] [PubMed]
  63. Seok, J.; Park, H.; Choi, J.H.; Lim, J.-Y.; Kim, K.G.; Kim, G.J. Placenta-derived mesenchymal stem cells restore the ovary function in an ovariectomized rat model via an antioxidant effect. Antioxidants 2020, 9, 591. [Google Scholar] [CrossRef]
  64. 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, 233–241. [Google Scholar] [CrossRef] [Green Version]
  65. Yang, Z.; Du, X.; Wang, C.; Zhang, J.; Liu, C.; Li, Y.; Jiang, H. Therapeutic effects of human umbilical cord mesenchymal stem cell-derived microvesicles on premature ovarian insufficiency in mice. Stem Cell Res. Ther. 2019, 10, 250. [Google Scholar] [CrossRef] [Green Version]
  66. Sun, L.; Li, D.; Song, K.; Wei, J.; Yao, S.; Li, Z.; Su, X.; Ju, X.; Chao, L.; Deng, X. 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. [Google Scholar] [CrossRef] [Green Version]
  67. Hong, L.; Yan, L.; Xin, Z.; Hao, J.; Liu, W.; Wang, S.; Liao, S.; Wang, H.; Yang, X. Protective effects of human umbilical cord mesenchymal stem cell-derived conditioned medium on ovarian damage. J. Mol. Cell Biol. 2020, 12, 372–385. [Google Scholar] [CrossRef]
  68. Zhang, J.; Yin, H.; Jiang, H.; Du, X.; Yang, Z. The protective effects of human umbilical cord mesenchymal stem cell-derived extracellular vesicles on cisplatin-damaged granulosa cells. Taiwan. J. Obstet. Gynecol. 2020, 59, 527–533. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, Q.; Bu, S.; Sun, J.; Xu, M.; Yao, X.; He, K.; Lai, D. Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage. Stem Cell Res. Ther. 2017, 8, 270. [Google Scholar] [CrossRef] [Green Version]
  70. Sun, B.; Ma, Y.; Wang, F.; Hu, L.; Sun, Y. miR-644-5p carried by bone mesenchymal stem cell-derived exosomes targets regulation of p53 to inhibit ovarian granulosa cell apoptosis. Stem Cell Res. Ther. 2019, 10, 360. [Google Scholar] [CrossRef] [PubMed]
  71. Yang, M.; Lin, L.; Sha, C.; Li, T.; Zhao, D.; Wei, H.; Chen, Q.; Liu, Y.; Chen, X.; Xu, W. 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, 342–352. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, Q.; Sun, J.; Huang, Y.; Bu, S.; Guo, Y.; Gu, T.; Li, B.; Wang, C.; Lai, D. Human amniotic epithelial cell-derived exosomes restore ovarian function by transferring microRNAs against apoptosis. Mol. Ther.-Nucleic Acids 2019, 16, 407–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ding, C.; Qian, C.; Hou, S.; Lu, J.; Zou, Q.; Li, H.; Huang, B. Exosomal miRNA-320a is released from hAMSCs and regulates SIRT4 to prevent reactive oxygen species generation in POI. Mol. Ther.-Nucleic Acids 2020, 21, 37–50. [Google Scholar] [CrossRef]
  74. Shi, Z.; Wang, Q.; Zhang, Y.; Jiang, D. Extracellular vesicles produced by bone marrow mesenchymal stem cells attenuate renal fibrosis, in part by inhibiting the RhoA/ROCK pathway, in a UUO rat model. Stem Cell Res. Ther. 2020, 11, 253. [Google Scholar] [CrossRef]
  75. Cacciottolo, M.; Morgan, T.E.; Saffari, A.A.; Shirmohammadi, F.; Forman, H.J.; Sioutas, C.; Finch, C.E. Traffic-related air pollutants (TRAP-PM) promote neuronal amyloidogenesis through oxidative damage to lipid rafts. Free Radic. Biol. Med. 2020, 147, 242–251. [Google Scholar] [CrossRef] [PubMed]
  76. Patel, S.; Korytkowski, M. Polycystic ovary syndrome: How best to establish the diagnosis. Women’s Health Prim. Care 2000, 3, 55–58. [Google Scholar]
  77. Franks, S.; White, D.M. Prevalence of and etiological factors in polycystic ovarian syndrome. Ann. N. Y. Acad. Sci. 1993, 687, 112–114. [Google Scholar] [CrossRef] [PubMed]
  78. Eid, G.M.; Cottam, D.R.; Velcu, L.M.; Mattar, S.G.; Korytkowski, M.T.; Gosman, G.; Hindi, P.; Schauer, P.R. Effective treatment of polycystic ovarian syndrome with Roux-en-Y gastric bypass. Surg. Obes. Relat. Dis. 2005, 1, 77–80. [Google Scholar] [CrossRef] [PubMed]
  79. Talbott, E.; Guzick, D.; Clerici, A.; Berga, S.; Detre, K.; Weimer, K.; Kuller, L. Coronary heart disease risk factors in women with polycystic ovary syndrome. Arterioscler. Thromb. Vasc. Biol. 1995, 15, 821–826. [Google Scholar] [CrossRef]
  80. Talbott, E.; Clerici, A.; Berga, S.L.; Kuller, L.; Guzick, D.; Detre, K.; Daniels, T.; Engberg, R.A. Adverse lipid and coronary heart disease risk profiles in young women with polycystic ovary syndrome: Results of a case-control study. J. Clin. Epidemiol. 1998, 51, 415–422. [Google Scholar] [CrossRef]
  81. Parham, K.J. Patient presentations: Panel discussion: Adenocarcinoma of the endometrium associated with the Stein-Leventhal syndrome. Am. J. Obstet. Gynecol. 1969, 105, 113–115. [Google Scholar] [CrossRef]
  82. Stein, I.F. Amenorrhea associated with bilateral polycystic ovaries. Am. J. Obs. Gynecol. 1935, 29, 181–191. [Google Scholar] [CrossRef]
  83. Harwood, K.; Vuguin, P.; DiMartino-Nardi, J. Current approaches to the diagnosis and treatment of polycystic ovarian syndrome in youth. Horm. Res. Paediatr. 2007, 68, 209–217. [Google Scholar] [CrossRef] [Green Version]
  84. Xie, Q.; Xiong, X.; Xiao, N.; He, K.; Chen, M.; Peng, J.; Su, X.; Mei, H.; Dai, Y.; Wei, D. Mesenchymal stem cells alleviate DHEA-Induced polycystic ovary syndrome (PCOS) by inhibiting inflammation in mice. Stem Cells Int. 2019, 2019, 9782373. [Google Scholar] [CrossRef] [Green Version]
  85. Zhao, Y.; Tao, M.; Wei, M.; Du, S.; Wang, H.; Wang, X. Mesenchymal stem cells derived exosomal miR-323-3p promotes proliferation and inhibits apoptosis of cumulus cells in polycystic ovary syndrome (PCOS). Artif. Cells Nanomed. Biotechnol. 2019, 47, 3804–3813. [Google Scholar] [CrossRef] [PubMed]
  86. Gargett, C.E. Uterine stem cells: What is the evidence? Hum. Reprod. Update 2007, 13, 87–101. [Google Scholar] [CrossRef]
  87. Jabbour, H.N.; Kelly, R.W.; Fraser, H.M.; Critchley, H.O. Endocrine regulation of menstruation. Endocr. Rev. 2006, 27, 17–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Zuo, W.; Xie, B.; Li, C.; Yan, Y.; Zhang, Y.; Liu, W.; Huang, J.; Chen, D. The clinical applications of endometrial mesenchymal stem cells. Biopreserv. Biobank. 2018, 16, 158–164. [Google Scholar] [CrossRef] [PubMed]
  89. Cervello, I.; Mas, A.; Gil-Sanchis, C.; Peris, L.; Faus, A.; Saunders, P.T.; Critchley, H.O.; Simon, C. Reconstruction of endometrium from human endometrial side population cell lines. PLoS ONE 2011, 6, e21221. [Google Scholar] [CrossRef] [PubMed]
  90. Gargett, C.E.; Schwab, K.E.; Zillwood, R.M.; Nguyen, H.P.; Wu, D. Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biol. Reprod. 2009, 80, 1136–1145. [Google Scholar] [CrossRef] [Green Version]
  91. Gargett, C.E.; Nguyen, H.P.; Ye, L. Endometrial regeneration and endometrial stem/progenitor cells. Rev. Endocr. Metab. Disord. 2012, 13, 235–251. [Google Scholar] [CrossRef]
  92. Ding, L.; Sun, H.; Su, J.; Lin, N.; Péault, B.; Song, T.; Yang, J.; Dai, J.; Hu, Y. Transplantation of bone marrow mesenchymal stem cells on collagen scaffolds for the functional regeneration of injured rat uterus. Biomaterials 2014, 35, 4888–4900. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, L.; Li, Y.; Guan, C.Y.; Tian, S.; Lv, X.D.; Li, J.-H.; Ma, X.; Xia, H.-F. Therapeutic effect of human umbilical cord-derived mesenchymal stem cells on injured rat endometrium during its chronic phase. Stem Cell Res. Ther. 2018, 9, 36. [Google Scholar] [CrossRef] [Green Version]
  94. Amable, P.R.; Carias, R.B.V.; Teixeira, M.V.T.; da Cruz Pacheco, Í.; do Amaral, R.J.F.C.; Granjeiro, J.M.; Borojevic, R. Platelet-rich plasma preparation for regenerative medicine: Optimization and quantification of cytokines and growth factors. Stem Cell Res. Ther. 2013, 4, 67. [Google Scholar] [CrossRef] [Green Version]
  95. Fotouhi, A.; Maleki, A.; Dolati, S.; Aghebati-Maleki, A.; Aghebati-Maleki, L. Platelet rich plasma, stromal vascular fraction and autologous conditioned serum in treatment of knee osteoarthritis. Biomed. Pharmacother. 2018, 104, 652–660. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, B.W.; Cheng, M.J.; Xu, C.J. Differentiation of mouse bone marrow mesenchymal stem cells into endometrial epithelial cells in vitro. Prog. Obstet. Gynecol. 2010, 19, 257–260. [Google Scholar]
  97. Yang, X.; Zhang, M.; Zhang, Y.; Li, W.; Yang, B. Mesenchymal stem cells derived from Wharton jelly of the human umbilical cord ameliorate damage to human endometrial stromal cells. Fertil. Steril. 2011, 96, 1029–1036.e1024. [Google Scholar] [CrossRef]
  98. Koippallil Gopalakrishnan, A.R.; Kishore, U.; Madan, T. Mesenchymal stem cells: A promising tool for targeted gene therapy of endometriosis. Regen. Med. 2017, 12, 69–76. [Google Scholar] [CrossRef] [Green Version]
  99. Xiao, B.; Zhu, Y.; Huang, J.; Wang, T.; Wang, F.; Sun, S. Exosomal transfer of bone marrow mesenchymal stem cell-derived miR-340 attenuates endometrial fibrosis. Biol. Open 2019, 8, bio039958. [Google Scholar] [CrossRef] [Green Version]
  100. Liang, L.; Wang, L.; Zhou, S.; Li, J.; Meng, L.; Zhang, H.; Cui, C.; Zhang, C. Exosomes derived from human umbilical cord mesenchymal stem cells repair injured endometrial epithelial cells. J. Assist. Reprod. Genet. 2020, 37, 395–403. [Google Scholar] [CrossRef]
  101. Wang, J.; Hu, R.; Xing, Q.; Feng, X.; Jiang, X.; Xu, Y.; Wei, Z. Exosomes Derived from Umbilical Cord Mesenchymal Stem Cells Alleviate Mifepristone-Induced Human Endometrial Stromal Cell Injury. Stem Cells Int. 2020, 2020, 6091269. [Google Scholar] [CrossRef] [PubMed]
  102. Senturk, L.M.; Erel, C.T. Thin endometrium in assisted reproductive technology. Curr. Opin. Obstet. Gynecol. 2008, 20, 221–228. [Google Scholar] [CrossRef]
  103. Chang, Y.; Li, J.; Chen, Y.; Wei, L.; Yang, X.; Shi, Y.; Liang, X. Autologous platelet-rich plasma promotes endometrial growth and improves pregnancy outcome during in vitro fertilization. Int. J. Clin. Exp. Med. 2015, 8, 1286. [Google Scholar] [PubMed]
  104. Tandulwadkar, S.R.; Naralkar, M.V.; Surana, A.D.; Selvakarthick, M.; Kharat, A.H. Autologous intrauterine platelet-rich plasma instillation for suboptimal endometrium in frozen embryo transfer cycles: A pilot study. J. Hum. Reprod. Sci. 2017, 10, 208. [Google Scholar] [CrossRef]
  105. Molina, A.; Sánchez, J.; Sánchez, W.; Vielma, V. Platelet-rich plasma as an adjuvant in the endometrial preparation of patients with refractory endometrium. JBRA Assist. Reprod. 2018, 22, 42. [Google Scholar] [CrossRef]
  106. Santamaria, X.; Cabanillas, S.; Cervello, I.; Arbona, C.; Raga, F.; Ferro, J.; Palmero, J.; Remohí, J.; Pellicer, A.; Simón, C. Autologous cell therapy with CD133+ bone marrow-derived stem cells for refractory Asherman’s syndrome and endometrial atrophy: A pilot cohort study. Hum. Reprod. 2016, 31, 1087–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Tersoglio, A.E.; Tersoglio, S.; Salatino, D.R.; Castro, M.; Gonzalez, A.; Hinojosa, M.; Castellano, O. Regenerative therapy by endometrial mesenchymal stem cells in thin endometrium with repeated implantation failure. A novel strategy. JBRA Assist. Reprod. 2020, 24, 118. [Google Scholar] [CrossRef]
  108. Jing, Z.; Qiong, Z.; Yonggang, W.; Yanping, L. Rat bone marrow mesenchymal stem cells improve regeneration of thin endometrium in rat. Fertil. Steril. 2014, 101, 587–594. [Google Scholar] [CrossRef]
  109. Simon, A.; Laufer, N. Assessment and treatment of repeated implantation failure (RIF). J. Assist. Reprod. Genet. 2012, 29, 1227–1239. [Google Scholar] [CrossRef] [Green Version]
  110. Achache, H.; Revel, A. Endometrial receptivity markers, the journey to successful embryo implantation. Hum. Reprod. Update 2006, 12, 731–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Fukui, A.; Funamizu, A.; Yokota, M.; Yamada, K.; Nakamua, R.; Fukuhara, R.; Kimura, H.; Mizunuma, H. Uterine and circulating natural killer cells and their roles in women with recurrent pregnancy loss, implantation failure and preeclampsia. J. Reprod. Immunol. 2011, 90, 105–110. [Google Scholar] [CrossRef] [PubMed]
  112. Gonen-Gross, T.; Goldman-Wohl, D.; Huppertz, B.; Lankry, D.; Greenfield, C.; Natanson-Yaron, S.; Hamani, Y.; Gilad, R.; Yagel, S.; Mandelboim, O. Inhibitory NK receptor recognition of HLA-G: Regulation by contact residues and by cell specific expression at the fetal-maternal interface. PLoS ONE 2010, 5, e8941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Asghari, S.; Valizadeh, A.; Aghebati-Maleki, L.; Nouri, M.; Yousefi, M. Endometriosis: Perspective, lights, and shadows of etiology. Biomed. Pharmacother. 2018, 106, 163–174. [Google Scholar] [CrossRef]
  114. Abdolmohammadi-Vahid, S.; Pashazadeh, F.; Pourmoghaddam, Z.; Aghebati-Maleki, L.; Abdollahi-Fard, S.; Yousefi, M. The effectiveness of IVIG therapy in pregnancy and live birth rate of women with recurrent implantation failure (RIF): A systematic review and meta-analysis. J. Reprod. Immunol. 2019, 134, 28–33. [Google Scholar] [CrossRef]
  115. Practice Committee of the American Society for Reproductive Medicine. Definitions of infertility and recurrent pregnancy loss: A committee opinion. Fertil. Steril. 2013, 99, 63. [Google Scholar] [CrossRef]
  116. Pourakbari, R.; Ahmadi, H.; Yousefi, M.; Aghebati-Maleki, L. Cell therapy in female infertility-related diseases: Emphasis on recurrent miscarriage and repeated implantation failure. Life Sci. 2020, 258, 118181. [Google Scholar] [CrossRef]
  117. Yu, N.; Zhang, B.; Xu, M.; Wang, S.; Liu, R.; Wu, J.; Yang, J.; Feng, L. Intrauterine administration of autologous peripheral blood mononuclear cells (PBMC s) activated by HCG improves the implantation and pregnancy rates in patients with repeated implantation failure: A prospective randomized study. Am. J. Reprod. Immunol. 2016, 76, 212–216. [Google Scholar] [CrossRef] [PubMed]
  118. Okitsu, O.; Kiyokawa, M.; Oda, T.; Miyake, K.; Sato, Y.; Fujiwara, H. Intrauterine administration of autologous peripheral blood mononuclear cells increases clinical pregnancy rates in frozen/thawed embryo transfer cycles of patients with repeated implantation failure. J. Reprod. Immunol. 2011, 92, 82–87. [Google Scholar] [CrossRef] [PubMed]
  119. Yoshioka, S.; Fujiwara, H.; Nakayama, T.; Kosaka, K.; Mori, T.; Fujii, S. Intrauterine administration of autologous peripheral blood mononuclear cells promotes implantation rates in patients with repeated failure of IVF–embryo transfer. Hum. Reprod. 2006, 21, 3290–3294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Hashii, K.; Fujiwara, H.; Yoshioka, S.; Kataoka, N.; Yamada, S.; Hirano, T.; Mori, T.; Fujii, S.; Maeda, M. Peripheral blood mononuclear cells stimulate progesterone production by luteal cells derived from pregnant and non-pregnant women: Possible involvement of interleukin-4 and interleukin-10 in corpus luteum function and differentiation. Hum. Reprod. (Oxf. Engl.) 1998, 13, 2738–2744. [Google Scholar] [CrossRef] [Green Version]
  121. Jensen, F.; Muzzio, D.; Soldati, R.; Fest, S.; Zenclussen, A.C. Regulatory B10 cells restore pregnancy tolerance in a mouse model. Biol. Reprod. 2013, 89, 90-1–90-7. [Google Scholar] [CrossRef] [Green Version]
  122. Gentile, T.; Borel, I.M.; Angelucci, J.; Miranda, S.; Margni, R.A. Preferential synthesis of asymmetric antibodies in rats immunized with paternal particulate antigens. Effect on pregnancy. J. Reprod. Immunol. 1992, 22, 173–183. [Google Scholar] [CrossRef]
  123. Sato, Y.; Fujiwara, H.; Zeng, B.-X.; Higuchi, T.; Yoshioka, S.; Fujii, S. Platelet-derived soluble factors induce human extravillous trophoblast migration and differentiation: Platelets are a possible regulator of trophoblast infiltration into maternal spiral arteries. Blood 2005, 106, 428–435. [Google Scholar] [CrossRef] [Green Version]
  124. Furukawa, K.; Fujiwara, H.; Sato, Y.; Zeng, B.-X.; Fujii, H.; Yoshioka, S.; Nishi, E.; Nishio, T. Platelets are novel regulators of neovascularization and luteinization during human corpus luteum formation. Endocrinology 2007, 148, 3056–3064. [Google Scholar] [CrossRef]
  125. Fujiwara, H. Immune cells contribute to systemic cross-talk between the embryo and mother during early pregnancy in cooperation with the endocrine system. Reprod. Med. Biol. 2006, 5, 19–29. [Google Scholar] [CrossRef] [Green Version]
  126. Metcalf, E. The effect of platelet-rich plasma (PRP) on intraluminal fluid and pregnancy rates in mares susceptible to persistent mating-induced endometritis (PMIE). J. Equine Vet. Sci. 2014, 1, 128. [Google Scholar] [CrossRef]
  127. Zadehmodarres, S.; Salehpour, S.; Saharkhiz, N.; Nazari, L. Treatment of thin endometrium with autologous platelet-rich plasma: A pilot study. JBRA Assist. Reprod. 2017, 21, 54. [Google Scholar] [CrossRef]
  128. Sadighi-Moghaddam, B.; Salek Farrokhi, A.; Namdar Ahmadabad, H.; Barati, M.; Moazzeni, S.M. Mesenchymal stem cell therapy prevents abortion in CBA/J× DBA/2 mating. Reprod. Sci. 2018, 25, 1261–1269. [Google Scholar] [CrossRef] [PubMed]
  129. Dmowski, W.P.; Greenblatt, R.B. Asherman’s syndrome and risk of placenta accreta. Obstet. Gynecol. 1969, 34, 288–299. [Google Scholar]
  130. Chirputkar, R.; Vaidya, A. Understanding Infertility and the Potential Role of Stem Cells in Infertility Treatment: A Short Com-munication. Int. J. Reprod. Fertil. Sex Health 2015, 2, 37–40. [Google Scholar]
  131. Yu, D.; Wong, Y.; Cheong, Y.; Xia, E.; Li, T.C. Asherman syndrome—one century later. Fertil. Steril. 2008, 89, 759–779. [Google Scholar] [CrossRef]
  132. March, C.M. Asherman’s Syndrome. Semin. Reprod. Med. 2011, 29, 83–94. [Google Scholar] [CrossRef]
  133. Lee, S.Y.; Shin, J.E.; Kwon, H.; Choi, D.H.; Kim, J.H. Effect of autologous adipose-derived stromal vascular fraction transplantation on endometrial regeneration in patients of Asherman’s syndrome: A pilot study. Reprod. Sci. 2020, 27, 561–568. [Google Scholar] [CrossRef]
  134. Singh, N.; Mohanty, S.; Seth, T.; Shankar, M.; Bhaskaran, S.; Dharmendra, S. Autologous stem cell transplantation in refractory Asherman’s syndrome: A novel cell based therapy. J. Hum. Reprod. Sci. 2014, 7, 93. [Google Scholar] [CrossRef]
  135. Nagori, C.B.; Panchal, S.Y.; Patel, H. Endometrial regeneration using autologous adult stem cells followed by conception by in vitro fertilization in a patient of severe Asherman’s syndrome. J. Hum. Reprod. Sci. 2011, 4, 43–48. [Google Scholar]
  136. Tan, J.; Li, P.; Wang, Q.; Li, Y.; Li, X.; Zhao, D.; Xu, X.; Kong, L. Autologous menstrual blood-derived stromal cells transplantation for severe Asherman’s syndrome. Hum. Reprod. 2016, 31, 2723–2729. [Google Scholar] [CrossRef] [Green Version]
  137. Zhao, S.; Qi, W.; Zheng, J.; Tian, Y.; Qi, X.; Kong, D.; Zhang, J.; Huang, X. Exosomes derived from adipose mesenchymal stem cells restore functional endometrium in a rat model of intrauterine adhesions. Reprod. Sci. 2020, 27, 1266–1275. [Google Scholar] [CrossRef]
  138. Xin, L.; Lin, X.; Zhou, F.; Li, C.; Wang, X.; Yu, H.; Pan, Y.; Fei, H.; Ma, L.; Zhang, S. A scaffold laden with mesenchymal stem cell-derived exosomes for promoting endometrium regeneration and fertility restoration through macrophage immunomodulation. Acta Biomater. 2020, 113, 252–266. [Google Scholar] [CrossRef]
  139. Yao, Y.; Chen, R.; Wang, G.; Zhang, Y.; Liu, F. Exosomes derived from mesenchymal stem cells reverse EMT via TGF-β1/Smad pathway and promote repair of damaged endometrium. Stem Cell Res. Ther. 2019, 10, 225. [Google Scholar] [CrossRef]
  140. Saribas, G.S.; Ozogul, C.; Tiryaki, M.; Pinarli, F.A.; Kilic, S.H. Effects of uterus derived mesenchymal stem cells and their exosomes on Asherman’s syndrome. Acta Histochem. 2020, 122, 151465. [Google Scholar] [CrossRef]
  141. Perrini, C.; Strillacci, M.G.; Bagnato, A.; Esposti, P.; Marini, M.G.; Corradetti, B.; Bizzaro, D.; Idda, A.; Ledda, S.; Capra, E. Microvesicles secreted from equine amniotic-derived cells and their potential role in reducing inflammation in endometrial cells in an in-vitro model. Stem Cell Res. Ther. 2016, 7, 169. [Google Scholar] [CrossRef] [Green Version]
  142. Lin, Y.-H.; Chen, Y.-H.; Chang, H.-Y.; Au, H.-K.; Tzeng, C.-R.; Huang, Y.-H. Chronic niche inflammation in endometriosis-associated infertility: Current understanding and future therapeutic strategies. Int. J. Mol. Sci. 2018, 19, 2385. [Google Scholar] [CrossRef] [Green Version]
  143. Parasar, P.; Ozcan, P.; Terry, K.L. Endometriosis: Epidemiology, diagnosis and clinical management. Curr. Obstet. Gynecol. Rep. 2017, 6, 34–41. [Google Scholar] [CrossRef] [Green Version]
  144. Bungum, H.F.; Vestergaard, C.; Knudsen, U.B. Endometriosis and type 1 allergies/immediate type hypersensitivity: A systematic review. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014, 179, 209–215. [Google Scholar] [CrossRef]
  145. Nielsen, N.M.; Jørgensen, K.T.; Pedersen, B.V.; Rostgaard, K.; Frisch, M. The co-occurrence of endometriosis with multiple sclerosis, systemic lupus erythematosus and Sjögren syndrome. Hum. Reprod. 2011, 26, 1555–1559. [Google Scholar] [CrossRef] [Green Version]
  146. Mu, F.; Rich-Edwards, J.; Rimm, E.B.; Spiegelman, D.; Forman, J.P.; Missmer, S.A. Association between endometriosis and hypercholesterolemia or hypertension. Hypertension 2017, 70, 59–65. [Google Scholar] [CrossRef]
  147. Kok, V.C.; Tsai, H.-J.; Su, C.-F.; Lee, C.-K. The risks for ovarian, endometrial, breast, colorectal, and other cancers in women with newly diagnosed endometriosis or adenomyosis: A population-based study. Int. J. Gynecol. Cancer 2015, 25, 968–976. [Google Scholar] [CrossRef]
  148. Králíčková, M.; Vetvicka, V. Endometriosis and ovarian cancer. World J. Clin. Oncol. 2014, 5, 800. [Google Scholar] [CrossRef]
  149. Zhao, Y.-x.; Chen, S.-r.; Su, P.-p.; Huang, F.-h.; Shi, Y.-c.; Shi, Q.-y.; Lin, S. Using mesenchymal stem cells to treat female infertility: An update on female reproductive diseases. Stem Cells Int. 2019, 2019, 9071720. [Google Scholar] [CrossRef]
  150. Vercellini, P.; Viganò, P.; Somigliana, E.; Fedele, L. Endometriosis: Pathogenesis and treatment. Nat. Rev. Endocrinol. 2014, 10, 261. [Google Scholar] [CrossRef]
  151. Maruyama, T.; Yoshimura, Y. Stem cell theory for the pathogenesis of endometriosis. Front. Biosci. 2012, 4, 2754–2763. [Google Scholar] [CrossRef] [Green Version]
  152. Serdar, E.; Bulun, E. Mechanisms of Disease. N. Engl. J. Med. 2009, 360, 268–279. [Google Scholar]
  153. Dunselman, G.; Vermeulen, N.; Becker, C.; Calhaz-Jorge, C.; D’Hooghe, T.; De Bie, B.; Heikinheimo, O.; Horne, A.; Kiesel, L.; Nap, A. ESHRE guideline: Management of women with endometriosis. Hum. Reprod. 2014, 29, 400–412. [Google Scholar] [CrossRef]
  154. Liu, Y.; Zhang, Z.; Yang, F.; Wang, H.; Liang, S.; Wang, H.; Yang, J.; Lin, J. The role of endometrial stem cells in the pathogenesis of endometriosis and their application to its early diagnosis. Biol. Reprod. 2020, 102, 1153–1159. [Google Scholar] [CrossRef] [PubMed]
  155. Simoni, M.; Taylor, H.S. Therapeutic strategies involving uterine stem cells in reproductive medicine. Curr. Opin. Obstet. Gynecol. 2018, 30, 209–216. [Google Scholar] [CrossRef]
  156. PAIN, O.P. Treatment of pelvic pain associated with endometriosis: A committee opinion. Fertil. Steril. 2014, 101, 927–935. [Google Scholar]
  157. Johnson, N.P.; Hummelshoj, L.; Consortium, W.E.S.M.; Abrao, M.; Adamson, G.; Allaire, C.; Amelung, V.; Andersson, E.; Becker, C.; Birna Árdal, K. Consensus on current management of endometriosis. Hum. Reprod. 2013, 28, 1552–1568. [Google Scholar] [CrossRef] [PubMed]
  158. Zito, G.; Luppi, S.; Giolo, E.; Martinelli, M.; Venturin, I.; Di Lorenzo, G.; Ricci, G. Medical treatments for endometriosis-associated pelvic pain. BioMed. Res. Int. 2014, 2014, 191967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Gopalakrishnan, A.K.; Pandit, H.; Metkari, S.; Warty, N.; Madan, T. Adenoviral vector encoding soluble Flt-1 engineered human endometrial mesenchymal stem cells effectively regress endometriotic lesions in NOD/SCID mice. Gene Ther. 2016, 23, 580–591. [Google Scholar] [CrossRef]
  160. Hajazimian, S.; Maleki, M.; Mehrabad, S.D.; Isazadeh, A. Human Wharton’s jelly stem cells inhibit endometriosis through apoptosis induction. Reproduction 2020, 159, 549–558. [Google Scholar] [CrossRef]
  161. LA, E. Problems in the diagnosis of adenomyosis uteri; with special reference to dysfunctional bleeding. West. J. Surg. Obstet. Gynecol. 1956, 64, 291–304, discussion, 304. [Google Scholar]
  162. Ferenczy, A. Pathophysiology of adenomyosis. Hum. Reprod. Update 1998, 4, 312–322. [Google Scholar] [CrossRef] [Green Version]
  163. Israel, L.S.; Woutersz, T.B. Adenomyosis: A neglected diagnosis. Obstet. Gynecol. 1959, 14, 168–173. [Google Scholar]
  164. Wood, C. Surgical and medical treatment of adenomyosis. Hum. Reprod. Update 1998, 4, 323–336. [Google Scholar] [CrossRef] [Green Version]
  165. Garcia, L.; Isaacson, K. Adenomyosis: Review of the literature. J. Minim. Invasive Gynecol. 2011, 18, 428–437. [Google Scholar] [CrossRef]
  166. Kim, M.D.; Kim, S.; Kim, N.K.; Lee, M.H.; Ahn, E.H.; Kim, H.J.; Cho, J.H.; Cha, S.H. Long-term results of uterine artery embolization for symptomatic adenomyosis. Am. J. Roentgenol. 2007, 188, 176–181. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, P.-H.; Liu, W.-M.; Fuh, J.-L.; Cheng, M.-H.; Chao, H.-T. Comparison of surgery alone and combined surgical-medical treatment in the management of symptomatic uterine adenomyoma. Fertil. Steril. 2009, 92, 876–885. [Google Scholar] [CrossRef] [PubMed]
  168. Phillips, D.R.; Nathanson, H.G.; Milim, S.J.; Haselkorn, J.S. Laparoscopic bipolar coagulation for the conservative treatment of adenomyomata. J. Am. Assoc. Gynecol. Laparosc. 1996, 4, 19–24. [Google Scholar] [CrossRef]
  169. Levgur, M. Therapeutic options for adenomyosis: A review. Arch. Gynecol. Obstet. 2007, 276, 1–15. [Google Scholar] [CrossRef] [PubMed]
  170. Fujishita, A.; Masuzaki, H.; Khan, K.N.; Kitajima, M.; Ishimaru, T. Modified reduction surgery for adenomyosis. Gynecol. Obstet. Investig. 2004, 57, 132–138. [Google Scholar] [CrossRef] [PubMed]
  171. Chen, Y.-J.; Li, H.-Y.; Chang, Y.-L.; Yuan, C.-C.; Tai, L.-K.; Lu, K.H.; Chang, C.-M.; Chiou, S.-H. Suppression of migratory/invasive ability and induction of apoptosis in adenomyo-sis-derived mesenchymal stem cells by cyclooxygenase-2 inhibitors. Fertil. Steril. 2010, 94, 1972–1979. [Google Scholar] [CrossRef]
  172. World Health Organisation. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction; Cambridge University Press: Cambridge, MA, USA, 1999. [Google Scholar]
  173. Harris, I.D.; Fronczak, C.; Roth, L.; Meacham, R.B. Fertility and the aging male. Rev. Urol. 2011, 13, e184. [Google Scholar]
  174. Tüttelmann, F. Classification of andrological disorders. In Andrology: Male Reproductive Health and Dysfunction; Nieschlag, E., Behre, H.M., Nieschlag, S., Eds.; Springer: Berlin/Heidelberg, Germany, 2010; Volume 4, pp. 87–92. [Google Scholar]
  175. Lipshultz, L.I.; Howards, S.S.; Niederberger, C.S. Infertility in the Male; Cambridge University Press: Cambridge, MA, USA, 2009. [Google Scholar]
  176. Irvine, D.S. Epidemiology and aetiology of male infertility. Hum. Reprod. 1998, 13, 33–44. [Google Scholar] [CrossRef] [Green Version]
  177. Zhang, D.; Liu, X.; Peng, J.; He, D.; Lin, T.; Zhu, J.; Li, X.; Zhang, Y.; Wei, G. Potential spermatogenesis recovery with bone marrow mesenchymal stem cells in an azoospermic rat model. Int. J. Mol. Sci. 2014, 15, 13151–13165. [Google Scholar] [CrossRef]
  178. Blackhall, F.H.; Atkinson, A.; Maaya, M.; Ryder, W.D.J.; Horne, G.; Brison, D.R.; Lieberman, B.; Radford, J.A. Semen cryopreservation, utilisation and reproductive outcome in men treated for Hodgkin’s disease. Br. J. Cancer 2002, 87, 381–384. [Google Scholar] [CrossRef] [Green Version]
  179. Fang, F.; Li, Z.; Zhao, Q.; Li, H.; Xiong, C. Human induced pluripotent stem cells and male infertility: An overview of current progress and perspectives. Hum. Reprod. 2018, 33, 188–195. [Google Scholar] [CrossRef]
  180. Esteves, S.C.; Miyaoka, R.; Agarwal, A. Surgical treatment of male infertility in the era of intracytoplasmic sperm injection-new insights. Clinics 2011, 66, 1463–1478. [Google Scholar] [CrossRef] [Green Version]
  181. Qamar, A.Y.; Fang, X.; Kim, M.J.; Cho, J. Improved viability and fertility of frozen-thawed dog sperm using adipose-derived mesenchymal stem cells. Sci. Rep. 2020, 10, 7034. [Google Scholar] [CrossRef]
  182. Qamar, A.Y.; Fang, X.; Kim, M.J.; Cho, J. Improved post-thaw quality of canine semen after treatment with exosomes from conditioned medium of adipose-derived mesenchymal stem cells. Animals 2019, 9, 865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Ghasemzadeh-Hasankolaei, M.; Eslaminejad, M.B.; Sedighi-Gilani, M. Derivation of male germ cells from ram bone marrow mesenchymal stem cells by three different methods and evaluation of their fate after transplantation into the testis. Vitr. Cell. Dev. Biol.-Anim. 2016, 52, 49–61. [Google Scholar] [CrossRef] [PubMed]
  184. Luo, Y.; Xie, L.; Mohsin, A.; Ahmed, W.; Xu, C.; Peng, Y.; Hang, H.; Zhuang, Y.; Chu, J.; Guo, M. Efficient generation of male germ-like cells derived during co-culturing of adipose-derived mesenchymal stem cells with Sertoli cells under retinoic acid and testosterone induction. Stem Cell Res. Ther. 2019, 10, 91. [Google Scholar] [CrossRef] [PubMed]
  185. Karimaghai, N.; Tamadon, A.; Rahmanifar, F.; Mehrabani, D.; Jahromi, A.R.; Zare, S.; Khodabandeh, Z.; Jahromi, I.R.; Koohi-Hoseinabadi, O.; Dianatpour, M. Spermatogenesis after transplantation of adipose tissue-derived mesenchymal stem cells in busulfan-induced azoospermic hamster. Iran. J. Basic Med. Sci. 2018, 21, 660–667. [Google Scholar]
  186. Tamadon, A.; Mehrabani, D.; Rahmanifar, F.; Jahromi, A.R.; Panahi, M.; Zare, S.; Khodabandeh, Z.; Jahromi, I.R.; Tanideh, N.; Dianatpour, M. Induction of spermatogenesis by bone marrow-derived mesenchymal stem cells in busulfan-induced azoospermia in hamster. Int. J. Stem Cells 2015, 8, 134. [Google Scholar] [CrossRef] [Green Version]
  187. Hou, J.; Yang, S.; Yang, H.; Liu, Y.; Liu, Y.; Hai, Y.; Chen, Z.; Guo, Y.; Gong, Y.; Gao, W.-Q. Generation of male differentiated germ cells from various types of stem cells. Reproduction 2014, 147, R179–R188. [Google Scholar] [CrossRef] [Green Version]
  188. Liu, L.; Banks, S.M.; Barnes, K.M.; Sherins, R.J. Two-year comparison of testicular responses to pulsatile gonadotropin-releasing hormone and exogenous gonadotropins from the inception of therapy in men with isolated hypogonadotropic hypogonadism. J. Clin. Endocrinol. Metab. 1988, 67, 1140–1145. [Google Scholar] [CrossRef] [PubMed]
  189. Rohayem, J.; Hauffa, B.P.; Zacharin, M.; Kliesch, S.; Zitzmann, M.; German Adolescent Hypogonadotropic Hypogonadism Study Group; Wüsthof, A.; Tittel, B.; Tittel, B.; Morlot, M. Testicular growth and spermatogenesis: New goals for pubertal hormone replacement in boys with hypogonadotropic hypogonadism? -a multicentre prospective study of hCG/rFSH treatment outcomes during adolescence-. Clin. Endocrinol. 2017, 86, 75–87. [Google Scholar] [CrossRef]
  190. Sani, R.N.; Tajik, P.; Yousefi, M.H.; Movahedin, M.; Qasemi-Panahi, B.; Shafiei, S.; Hamedani, M.A. Follicle stimulating hormone increases spermatogonial stem cell colonization during in vitro co-culture. In Veterinary Research Forum: An International Quarterly Journal; Faculty of Veterinary Medicine, Urmia University: Urmia, Iran, 2013; p. 37. [Google Scholar]
  191. Tajik, P.; Sani, R.N.; Moezifar, M.; Yousefi, M.; Movahedin, M.; Qasemi-Panahi, B.; Shafiei, S.; Fili, P.R. Effect of follicle-stimulating hormone and testosterone on colony formation of bovine spermatogonial stem cell. Comp. Clin. Pathol. 2014, 23, 901–906. [Google Scholar] [CrossRef]
  192. Su, J.S.; Farber, N.J.; Vij, S.C. Pathophysiology and treatment options of varicocele: An overview. Andrologia 2020, 53, e13576. [Google Scholar] [CrossRef]
  193. Agarwal, A.; Hamada, A.; Esteves, S.C. Insight into oxidative stress in varicocele-associated male infertility: Part 1. Nat. Rev. Urol. 2012, 9, 678. [Google Scholar] [CrossRef] [PubMed]
  194. Raheem, A.A.; Ralph, D. Male infertility: Causes and investigations. Trends Urol. Men’s Health 2011, 2, 8–11. [Google Scholar] [CrossRef]
  195. Goldstein, M.; Eid, J.-F. Elevation of intratesticular and scrotal skin surface temperature in men with varicocele. J. Urol. 1989, 142, 743–745. [Google Scholar] [CrossRef]
  196. Lipshultz, L.I.; Corriere, J.N., Jr. Progressive testicular atrophy in the varicocele patient. J. Urol. 1977, 117, 175–176. [Google Scholar] [CrossRef]
  197. Rashtbari, H.; Razi, M.; Hassani-Bafrani, H.; Najaran, H. Berberine reinforces Sertoli cells niche and accelerates spermatogonial stem cells renewal in experimentally-induced varicocele condition in rats. Phytomedicine 2018, 40, 68–78. [Google Scholar] [CrossRef]
  198. Liu, H.-C.; Xie, Y.; Deng, C.-H.; Liu, G.-H. Stem cell-based therapies for fertility preservation in males: Current status and future prospects. World J. Stem Cells 2020, 12, 1097. [Google Scholar] [CrossRef]
  199. Mohammad, S.; Aghamir, K.; Salavati, A.; Yousefie, R.; Tootian, Z.; Ghazaleh, N.; Jamali, M.; Azimi, P. Does Bone Marrow–derived Mesenchymal Stem Cell Transfusion Prevent Antisperm Antibody Production After Traumatic Testis Rupture? Urology 2014, 86, 82–86. [Google Scholar] [CrossRef]
  200. Addas, F.; Yan, S.; Hadjipavlou, M.; Gonsalves, M.; Sabbagh, S. Testicular Rupture or Testicular Fracture? A Case Report and Literature Review. Case Rep. Urol. 2018, 2018, 1323780. [Google Scholar] [CrossRef] [PubMed]
  201. Sreenivasa, G.; Kavitha, P.; Chaithra, P.; Vineeth, V.; Kumar, C.S.; Malini, S.S. Clinical Significance of antisperm antibody analysis in evaluating male infertility of south karnataka. Age 2011, 20, 31–40. [Google Scholar]
  202. Fahlbusch, B.; Fahlbusch, M.; Thon, W. Blunt testicular injury-conservative or surgical treatment? Aktuelle Urol. 2003, 34, 176–178. [Google Scholar] [PubMed]
  203. Haas, G.G., Jr.; Manganiello, P. A double-blind, placebo-controlled study of the use of methylprednisolone in infertile men with sperm-associated immunoglobulins. Fertil. Steril. 1987, 47, 295–301. [Google Scholar] [CrossRef]
  204. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  205. Abdi, R.; Fiorina, P.; Adra, C.N.; Atkinson, M.; Sayegh, M.H. Immunomodulation by mesenchymal stem cells: A potential therapeutic strategy for type 1 diabetes. Diabetes 2008, 57, 1759–1767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Corcione, A.; Benvenuto, F.; Ferretti, E.; Giunti, D.; Cappiello, V.; Cazzanti, F.; Risso, M.; Gualandi, F.; Mancardi, G.L.; Pistoia, V. Human mesenchymal stem cells modulate B-cell functions. Blood 2006, 107, 367–372. [Google Scholar] [CrossRef] [Green Version]
  207. Hsiao, C.-H.; Ji, A.T.-Q.; Chang, C.-C.; Cheng, C.-J.; Lee, L.M.; Ho, J.H.-C. Local injection of mesenchymal stem cells protects testicular torsion-induced germ cell injury. Stem Cell Res. Ther. 2015, 6, 113. [Google Scholar] [CrossRef] [Green Version]
  208. Wilhelm Filho, D.; Torres, M.A.; Bordin, A.L.; Crezcynski-Pasa, T.B.; Boveris, A. Spermatic cord torsion, reactive oxygen and nitrogen species and ischemia–reperfusion injury. Mol. Asp. Med. 2004, 25, 199–210. [Google Scholar] [CrossRef] [PubMed]
  209. Reyes, J.G.; Farias, J.G.; Henríquez-Olavarrieta, S.; Madrid, E.; Parraga, M.; Zepeda, A.B.; Moreno, R.D. The hypoxic testicle: Physiology and pathophysiology. Oxidative Med. Cell. Longev. 2012, 2012, 929285. [Google Scholar] [CrossRef]
  210. Dokmeci, D. Testicular torsion, oxidative stress and the role of antioxidant therapy. Folia Med. 2006, 48, 16–21. [Google Scholar]
  211. Mansbach, J.M.; Forbes, P.; Peters, C. Article. Arch. Pediatrics Adolesc. Med. 2005, 159, 1167–1171. [Google Scholar] [CrossRef] [PubMed]
  212. Azizollahi, S.; Aflatoonian, R.; Gilani, M.A.S.; Behnam, B.; Tajik, N.; Asghari-Jafarabadi, M.; Reza Asgari, H.; Koruji, M. Alteration of spermatogenesis following spermatogonial stem cells transplantation in testicular torsion-detorsion mice. J. Assist. Reprod. Genet. 2016, 33, 771–781. [Google Scholar] [CrossRef] [Green Version]
  213. Hsiao, C.-H.; Ji, A.T.-Q.; Chang, C.-C.; Cheng, C.-J.; Lee, L.M.; Ho, J.H.-C. Mesenchymal stem cells restore the sperm motility from testicular torsion-detorsion injury by regulation of glucose metabolism in sperm. Stem Cell Res. Ther. 2019, 10, 270. [Google Scholar] [CrossRef]
  214. Zhong, L.; Yang, M.; Zou, X.; Du, T.; Xu, H.; Sun, J. Human umbilical cord multipotent mesenchymal stromal cells alleviate acute ischemia-reperfusion injury of spermatogenic cells via reducing inflammatory response and oxidative stress. Stem Cell Res. Ther. 2020, 11, 294. [Google Scholar] [CrossRef]
  215. Klinefelter, H.F., Jr.; Reifenstein, E.C., Jr.; Albright, F., Jr. Syndrome characterized by gynecomastia, aspermatogenesis without A-Leydigism, and increased excretion of follicle-stimulating hormone. J. Clin. Endocrinol. 1942, 2, 615–627. [Google Scholar] [CrossRef]
  216. Ferlin, A.; Arredi, B.; Foresta, C. Genetic causes of male infertility. Reprod. Toxicol. 2006, 22, 133–141. [Google Scholar] [CrossRef]
  217. Bojesen, A.; Juul, S.; Gravholt, C.H. Prenatal and postnatal prevalence of Klinefelter syndrome: A national registry study. J. Clin. Endocrinol. Metab. 2003, 88, 622–626. [Google Scholar] [CrossRef]
  218. Mehta, A.; Bolyakov, A.; Roosma, J.; Schlegel, P.N.; Paduch, D.A. Successful testicular sperm retrieval in adolescents with Klinefelter syndrome treated with at least 1 year of topical testosterone and aromatase inhibitor. Fertil. Steril. 2013, 100, 970–974. [Google Scholar] [CrossRef] [PubMed]
  219. Wyns, C.; Curaba, M.; Vanabelle, B.; Van Langendonckt, A.; Donnez, J. Options for fertility preservation in prepubertal boys. Hum. Reprod. Update 2010, 16, 312–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Song, K.-J.; Jeong, S.-M.; Park, S.-J.; Song, K.-H.; Choi, H.-J.; Lee, Y.-W.; Yoon, K.-Y.; Shin, S.-T.; Cho, S.-W.; Cho, J.-K. A Case of the Periorchiepididymitis in a Dog. J. Vet. Clin. 2008, 25, 40–43. [Google Scholar]
  221. Malolina, E.A.; Kulibin, A.Y.; Kushch, A.A. Neonatal testicular cell transplantation restores murine spermatogenesis damaged in the course of herpes simplex virus-induced orchitis. Reprod. Fertil. Dev. 2016, 28, 757–764. [Google Scholar] [CrossRef]
  222. Kaver, I.; Matzkin, H.; Braf, Z.F. Epididymo-orchitis: A retrospective study of 121 patients. J. Fam. Pract. 1990, 30, 548–553. [Google Scholar]
  223. Kadish, H.A.; Bolte, R.G. A retrospective review of pediatric patients with epididymitis, testicular torsion, and torsion of testicular appendages. Pediatrics 1998, 102, 73–76. [Google Scholar] [CrossRef]
  224. Luzzi, G.; O’brien, T. Acute epididymitis. BJU Int. 2001, 87, 747–755. [Google Scholar] [CrossRef]
  225. Thomas, T.H.; Lishnak, T.S.; Heiman, D. Epididymitis and orchitis: An overview. Am. Fam. Physician 2009, 79, 583–587. [Google Scholar]
  226. Ludwig, M. Diagnosis and therapy of acute prostatitis, epididymitis and orchitis. Andrologia 2008, 40, 76–80. [Google Scholar] [CrossRef] [PubMed]
  227. Manavi, K.; Turner, K.; Scott, G.; Stewart, L. Audit on the management of epididymo-orchitis by the Department of Urology in Edinburgh. Int. J. STD AIDS 2005, 16, 386–387. [Google Scholar] [CrossRef] [PubMed]
  228. Erpenbach, K.H. Systemic treatment with Interferon-α2b: An effective method to prevent sterility after bilateral mumps orchitis. J. Urol. 1991, 146, 54–56. [Google Scholar] [CrossRef]
  229. Ku, J.-H.; Yim, Y.-S.; Lee, C.-H.; Kim, Y.-H.; Song, Y.-S.; Jeon, Y.-S.; Kim, M.-E.; Lee, N.-K.; Park, Y.-H. The Preventive Effect of Systemic Treatment with Interferon-${\alpha} 2B $ for Infertility of Mumps Orchitis. Clin. Exp. Reprod. Med. 1999, 26, 111–116. [Google Scholar]
  230. Yeniyol, C.O.; Sorguc, S.; Minareci, S.; Ayder, A.R. Role of interferon-alpha-2B in prevention of testicular atrophy with unilateral mumps orchitis. Urology 2000, 55, 931–933. [Google Scholar] [CrossRef]
  231. Sherif, I.O.; Abdel-Aziz, A.; Sarhan, O.M. Cisplatin-induced testicular toxicity in rats: The protective effect of arjunolic acid. J. Biochem. Mol. Toxicol. 2014, 28, 515–521. [Google Scholar] [CrossRef]
  232. Awadalla, E.A. Ameliorative effect of the crude oil of the Nigella sativa on oxidative stress induced in rat testes by cisplatin treatment. Biomed. Prev. Nutr. 2012, 2, 265–268. [Google Scholar] [CrossRef]
  233. Jahnukainen, K.; Mitchell, R.T.; Stukenborg, J.-B. Testicular function and fertility preservation after treatment for haematological cancer. Curr. Opin. Endocrinol. Diabetes Obes. 2015, 22, 217–223. [Google Scholar] [CrossRef]
  234. Clermont, Y. Kinetics of spermatogenesis in mammals: Seminiferous epithelium cycle and spermatogonial renewal. Physiol. Rev. 1972, 52, 198–236. [Google Scholar] [CrossRef]
  235. De Michele, F.; Vermeulen, M.; Wyns, C. Fertility restoration with spermatogonial stem cells. Curr. Opin. Endocrinol. Diabetes Obes. 2017, 24, 424–431. [Google Scholar] [CrossRef] [PubMed]
  236. Wang, Y.-J.; Yan, J.; Zou, X.-L.; Guo, K.-J.; Zhao, Y.; Meng, C.-Y.; Yin, F.; Guo, L. Bone marrow mesenchymal stem cells repair cadmium-induced rat testis injury by inhibiting mitochondrial apoptosis. Chem.-Biol. Interact. 2017, 271, 39–47. [Google Scholar] [CrossRef] [PubMed]
  237. Sherif, I.O.; Sabry, D.; Abdel-Aziz, A.; Sarhan, O.M. The role of mesenchymal stem cells in chemotherapy-induced gonadotoxicity. Stem Cell Res. Ther. 2018, 9, 196. [Google Scholar] [CrossRef]
  238. Mohammed, F.Z.; Hassan Abdallah, S.; Hassan Watad, S. Treatment of cyclophosphamide induced infertile male mice with HSCs that homed by honey, bovine colostrum and umbilical cord blood derived mesenchymal stem cells. Biochem. Lett. 2019, 15, 98–113. [Google Scholar] [CrossRef]
  239. Abdelaziz, M.H.; Salah EL-Din, E.Y.; El-Dakdoky, M.H.; Ahmed, T.A. The impact of mesenchymal stem cells on doxorubicin-induced testicular toxicity and progeny outcome of male prepubertal rats. Birth Defects Res. 2019, 111, 906–919. [Google Scholar] [CrossRef] [PubMed]
  240. Cai, Y.-T.; Xiong, C.-L.; Liu, T.-S.; Shen, S.-L.; Rao, J.-P.; Qiu, F. Secretions released from mesenchymal stem cells improve spermatogenesis restoration of cytotoxic treatment with busulfan in azoospermia mice. Andrologia 2021, 53, e14144. [Google Scholar] [CrossRef]
  241. Cai, Y.t.; Xiong, C.l.; Shen, S.l.; Rao, J.p.; Liu, T.s.; Qiu, F. Mesenchymal stem cell-secreted factors delayed spermatogenesis injuries induced by busulfan involving intercellular adhesion molecule regulation. Andrologia 2019, 51, e13285. [Google Scholar] [CrossRef]
  242. Deng, C.; Xie, Y.; Zhang, C.; Ouyang, B.; Chen, H.; Lv, L.; Yao, J.; Liang, X.; Zhang, Y.; Sun, X. Urine-derived stem cells facilitate endogenous spermatogenesis restoration of busulfan-induced nonobstructive azoospermic mice by paracrine exosomes. Stem Cells Dev. 2019, 28, 1322–1333. [Google Scholar] [CrossRef]
  243. Mobarak, H.; Heidarpour, M.; Rahbarghazi, R.; Nouri, M.; Mahdipour, M. Amniotic fluid-derived exosomes improved spermatogenesis in a rat model of azoospermia. Life Sci. 2021, 274, 119336. [Google Scholar] [CrossRef]
  244. Zhou, Q.-Z.; Liu, C.-D.; Guo, X.-B.; Zhai, J.-W.; Xia, H.; Yang, J.-K.; Zhou, J.-H.; Guo, W.-B.; Yang, C.; Xia, M.; et al. Protective effect of bone marrow mesenchymal stem cell-derived exosomes against the reproductive toxicity of cyclophosphamide is associated with the p38MAPK/ERK and AKT signaling pathways. Asian J. Androl. 2021, 23, 386. [Google Scholar] [CrossRef]
  245. Salek, F.; Baharara, J.; Shahrokhabadi, K.N.; Amini, E. The guardians of germ cells; Sertoli-derived exosomes against electromagnetic field-induced oxidative stress in mouse spermatogonial stem cells. Theriogenology 2021, 173, 112–122. [Google Scholar] [CrossRef]
  246. Zhang, W.; Yang, C.; Guo, W.; Guo, X.; Bian, J.; Zhou, Q.; Chen, M.; Zhou, J.; Chen, Z.; Wang, P.; et al. Protective effect of bone marrow mesenchymal stem cells-derived exosomes against testicular ischemia-reperfusion injury in rats. Nan Fang Yi Ke Da Xue Xue Bao J. South. Med Univ. 2018, 38, 910–916. [Google Scholar] [CrossRef]
  247. Abdelaal, N.E.; Tanga, B.M.; Abdelgawad, M.; Allam, S.; Fathi, M.; Saadeldin, I.M.; Bang, S.; Cho, J. Cellular Therapy via Spermatogonial Stem Cells for Treating Impaired Spermatogenesis, Non-Obstructive Azoospermia. Cells 2021, 10, 1779. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of extracellular vesicles (EVs) uptake and cargo release to the target cells. EVs can fuse with the plasma membrane and deliver the molecular cargo into the cytoplasm. The EV surface protein can interact with cell receptors and deliver a signal pathway. EVs may be up-taken into the cells through pinocytosis where lysosomes can recycle or release their contents into the cytoplasm. The molecular contents of the EVs reflect the cell of origin and exhibit their actions when they become free from the lipid bilayer.
Figure 1. Schematic illustration of extracellular vesicles (EVs) uptake and cargo release to the target cells. EVs can fuse with the plasma membrane and deliver the molecular cargo into the cytoplasm. The EV surface protein can interact with cell receptors and deliver a signal pathway. EVs may be up-taken into the cells through pinocytosis where lysosomes can recycle or release their contents into the cytoplasm. The molecular contents of the EVs reflect the cell of origin and exhibit their actions when they become free from the lipid bilayer.
Cells 10 02460 g001
Figure 2. Diagrammatic illustration of the uses of stem cells (SCs) and their derived extracellular vesicles (EVs) for treating male and female infertility. Stem cells can be isolated from different sources such as cord blood, bone marrow, and adipose-derived stem cells (ADSCs) and used for culture and propagation. Cultured stem cells are used for isolating EVs from their conditioned medium. SCs and/or EVs can be administered to treat infertility diseases. POF: Premature ovarian failure. PCOS: Polycystic ovarian syndrome.
Figure 2. Diagrammatic illustration of the uses of stem cells (SCs) and their derived extracellular vesicles (EVs) for treating male and female infertility. Stem cells can be isolated from different sources such as cord blood, bone marrow, and adipose-derived stem cells (ADSCs) and used for culture and propagation. Cultured stem cells are used for isolating EVs from their conditioned medium. SCs and/or EVs can be administered to treat infertility diseases. POF: Premature ovarian failure. PCOS: Polycystic ovarian syndrome.
Cells 10 02460 g002
Table 1. Uses of stem cell-derived extracellular vesicles for treating female infertility.
Table 1. Uses of stem cell-derived extracellular vesicles for treating female infertility.
Female Infertility CategoryAffectionEvs Source/Route of Administration in VivoMain Mechanism of ActionsReference
Premature ovarian failurePremature ovarian insufficiency (in vivo, mice model)HUCMSCs/intravenous injectionUpregulation of AKT, p-AKT, and angiogenic cytokines (including VEGF, IGF, and angiogenin)[65]
Ovarian granulosa cell cisplatin-treated (in vitro, rats model)HUCMSCsBcl-2 and caspase-3 were upregulated, whilst the expression of Bax, cleaved caspase-3, and cleaved PARP were downregulated [66]
Ovarian granulosa cell cisplatin-treated (in vitro, rats model)HUCMSCsUpregulated E2, StAR, and Bcl-2/Bax ratio
downregulated Caspase 3
[68]
Premature ovarian insufficiency (in vivo, mice model) & human granulosa-lutein cells (in vitro)HAECs/intraovarian injection109 cytokines involved in apoptosis, angiogenesis, cell cycle and immune response (in vivo)
Decreasing apoptosis and TGF-β/Smad signaling pathway (in vitro)
[69]
Premature ovarian insufficiency (in vivo, mice model)BMSCs/intravenous injectionmiR-664-5p targeted p53 and apoptosis[70]
Premature ovarian insufficiency (in vivo, rats model)BMSCs/intraperitoneal injectionmiR-144-5p targeted PTEN and apoptosis[71]
Premature ovarian insufficiency (in vivo, mice model)HAECs/intraovarian injectionmiR-1246 targeted phosphatidylinositol signaling and apoptosis pathways[72]
Premature ovarian insufficiency (in vivo, mice model)AMSCs/intraovarian injectionmiR-320a targeted SIRT4 and ROS formation [73]
PCOSCumulus cells of PCOS patients (in vitro)Human AMSCsmiR-323-3p promoted cell proliferation and inhibited apoptosis in CCs through targeting PDCD4[85]
Endometrial injuriesRats subjected to mechanical endometrial damage BMSCs/intravenous injectionmiR-340 downregulated collagen 1α1, α-SMA and transforming growth factor (TGF)-β1[99]
Mice injured endometrial epithelial cells (in vitro)HUCMSCsInhibited IL-6, IL-1β, TLR4 and RelA
Increased TNFA
[100]
Human endometrial stromal cells injury (in vitro)HUCMSCsIncreasing Bcl-2 level and downregulating Cleaved Caspase-3 level and activated the PTEN/AKT signaling pathway to regulate the proliferation and antiapoptosis[101]
Asherman’s syndromeRat model (in vivo)ADMSCs/intrauterine injectionEnhanced the expression of integrin-β3, LIF, and VEGF[137]
Rat model (in vivo)HUCMSCs & collagen scaffold/endometrial transplantationmiRNAs facilitated CD163+ M2 macrophage polarization, reduced inflammation, and increased anti-inflammatory responses[138]
Rabbit model (in vivo)BMSCs/Uterine muscle injectionCK19 level significantly increased whereas VIM level significantly decreased
TGF-β1, TGF-β1R, and Smad2 mRNA were all significantly decreased
[139]
Rat model (in vivo)Uterus MSCs/intrauterine injectionMMP-2 and MMP-9 expression was enhanced
TIMP-2 expression was decreased
[140]
EndometriosisLipopolysaccharides effects on endometrial cell (equines, in vitro)Equine AMSCsReduced tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin 1β (IL-1β), and metalloproteinases (MMP) 1 and 13, and the release of some pro- or anti-inflammatory cytokines.[141]
UCMSCs: human umbilical cord mesenchymal stem cells, HAECs: human amniotic epithelial cells, BMSC: bone marrow mesenchymal stem cell, AMSCs: amniotic mesenchymal stem cells, ADMSCs: Adipose mesenchymal stem cells.
Table 2. Uses of stem cell-derived extracellular vesicles for treating male infertility caused by gonadotoxicity and non-obstructive azoospermia (NOA).
Table 2. Uses of stem cell-derived extracellular vesicles for treating male infertility caused by gonadotoxicity and non-obstructive azoospermia (NOA).
Model of DiseaseTherapeutic Intervention/Route of Administration Core FindingsReference
Busulfan-induced NOA mice modelUrine-derived stem cells -derived EVs/Intratesticularspermatogenic genes (Pou5f1, Prm1, SYCP3, and DAZL) and the spermatogenic protein UCHL1 were significantly increased after 36 days of injection[242]
Busulfan-induced NOA rats modelAmniotic fluid-derived EVs/IntratesticularDAZL and VASA were increased significantly.
Sperm parameters and spermatogenesis index were significantly improved.
OCT-3/4+ cells were increased in NOA rats after AF-Exos injection, showing the restoration of spermatogenesis.
[243]
Cyclophosphamide-induced testicular spermatogenic dysfunctionBone marrow mesenchymal stem cell-derived EVs/intravenousIncreased spermatogonia cell proliferation and reduced apoptosis. Phosphorylated levels of ERK, AKT, and p38MAPK proteins were reduced[244]
Electromagnetic field-induced oxidative stress in mouse spermatogonial stem cells (in vitro)Sertoli cells-derived EVsdown-regulation of the apoptotic gene (Caspase-3), and oxidative stress.
Up-regulation of SSCs specific gene (GFRα1).
[245]
Testicular ischemia-reperfusion injury in ratsBone marrow mesenchymal stem cell-derived EVs/IntratesticularReduced HMGB1, caspase-3, and cleaved caspase-3[246]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Qamar, A.Y.; Hussain, T.; Rafique, M.K.; Bang, S.; Tanga, B.M.; Seong, G.; Fang, X.; Saadeldin, I.M.; Cho, J. The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility. Cells 2021, 10, 2460. https://doi.org/10.3390/cells10092460

AMA Style

Qamar AY, Hussain T, Rafique MK, Bang S, Tanga BM, Seong G, Fang X, Saadeldin IM, Cho J. The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility. Cells. 2021; 10(9):2460. https://doi.org/10.3390/cells10092460

Chicago/Turabian Style

Qamar, Ahmad Yar, Tariq Hussain, Muhammad Kamran Rafique, Seonggyu Bang, Bereket Molla Tanga, Gyeonghwan Seong, Xun Fang, Islam M. Saadeldin, and Jongki Cho. 2021. "The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility" Cells 10, no. 9: 2460. https://doi.org/10.3390/cells10092460

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop