Cell therapy for the treatment of reproductive diseases and infertility: an overview from the mechanism to the clinic alongside diagnostic methods

Kosar Babaei , Mohsen Aziminezhad , Seyedeh Elham Norollahi , Sogand Vahidi , Ali Akbar Samadani

Front. Med. ›› 2022, Vol. 16 ›› Issue (6) : 827 -858.

PDF (3344KB)
Front. Med. ›› 2022, Vol. 16 ›› Issue (6) : 827 -858. DOI: 10.1007/s11684-022-0948-8
REVIEW
REVIEW

Cell therapy for the treatment of reproductive diseases and infertility: an overview from the mechanism to the clinic alongside diagnostic methods

Author information +
History +
PDF (3344KB)

Abstract

Infertility is experienced by 8%12% of adults in their reproductive period globally and has become a prevalent concern. Besides routine therapeutic methods, stem cells are rapidly being examined as viable alternative therapies in regenerative medicine and translational investigation. Remarkable progress has been made in understanding the biology and purpose of stem cells. The affected pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) are further studied for their possible use in reproductive medicine, particularly for infertility induced by premature ovarian insufficiency and azoospermia. Accordingly, this study discusses current developments in the use of some kinds of MSCs such as adipose-derived stem cells, bone marrow stromal cells, umbilical cord MSCs, and menstrual blood MSCs. These methods have been used to manage ovarian and uterine disorders, and each technique presents a novel method for the therapy of infertility.

Keywords

infertility / stem cell therapy / mesenchymal stem cells / pluripotent stem cells

Cite this article

Download citation ▾
Kosar Babaei, Mohsen Aziminezhad, Seyedeh Elham Norollahi, Sogand Vahidi, Ali Akbar Samadani. Cell therapy for the treatment of reproductive diseases and infertility: an overview from the mechanism to the clinic alongside diagnostic methods. Front. Med., 2022, 16(6): 827-858 DOI:10.1007/s11684-022-0948-8

登录浏览全文

4963

注册一个新账户 忘记密码

1 Introduction

Infertility is characterized by the inability to get pregnant for at least 12 months [1]. Infertility can be classified into three primary classifications, namely, female reasons, male reasons, and combined causes [2]. Female infertility is caused by various circumstances, such as disorders of the reproductive system. In this way, ovulation abnormalities (e.g., polycystic ovary syndrome, hypothalamus instability, and primary ovarian failure), tubal infertility, endometriosis, and uterine and cervical reasons cause female infertility (e.g., polyps, cervical stenosis, and tumors). Hormone replacement therapy can help with infertility in some cases, but previous studies have confirmed that it raises the breast cancer risk [3,4]. Although several reasons linked to increased risks for multiple pregnancies should be examined, ovulation induction, superovulation, or assisted reproductive technologies results showed tendencies toward increasing fertility rates [5] (Fig.1).

Remarkably, alternative treatments for infertility, including stem cell treatment, have been studied by researchers. Stem cells are undifferentiated cells with the potential to renew themselves for lengthy periods without major modifications in their general features. Under particular physiologic or experimental settings, they can develop into various specialized cells. A significant interest has been observed because of the constraints of employing embryonic and induced pluripotent stem cells in the therapy. One of the most important elements in this type of therapy is the use of mesenchymal stem cells (MSCs). Correspondingly, MSCs are devoid of both ethical problems and the production of teratomas [6]. MSCs, which are commonly known as mesenchymal stromal cells, are a type of pluripotent stem cell that originates in the mesoderm. They can self-renew and differentiate into ectodermic and endodermic cells and mesoderm lineages, including osteocytes, chondrocytes, and adipocytes [7,8]. Considerably, adult tissues, including the umbilical cord (UC), menstrual blood, adipose tissue (AD), placenta, and the bone marrow, can all be used to extract MSCs in the therapy [9,10]. Overall, hormonal imbalance and the physical and mental health are related to the difficulties associated with reproductive diseases and infertility. Correspondingly, assisted reproductive tools can only work out approximately 50% of the patients. Furthermore, it involves significant problems and does not resolve the basic difficulty of infertility. Considering that pluripotent stem cells can differentiate into almost any kind of different cells, they have been extremely associated as an optimal choice in the progress of stem cell-based fertility treatments, which could also regulate genetic disorders and diseases in all offspring. These developments in reproductive technology indicate both oppositions and feasibilities for solving infertility problems caused by different parameters. This paper comprehensively indicates and clarifies all types of infertility diseases and the potential use of stem cells for the treatment of these reproductive disorders.

2 Current therapeutic of infertility

Although therapeutic intervention has remarkably advanced in recent years, more than 80% of couples experiences infertility [11]. Infertility is a worldwide problem that affects over 15% of all couples, in which male reasons account for roughly 30% of cases, and female factors account for roughly 40% of situations [12]. Infertility is a complicated disease, and therapy varies depending on the age of the patient, the cause of infertility, and other factors. Importantly, therapies must be able to endure financial, temporal, physical, and psychological constraints (Tab.1).

Several therapies, including surgery and medicine, may be indicated at roughly the same time based on patient characteristics. Fertility medications are mainly used for promoting ovulation in women with ovulation problems. The ovarian follicles rupture, and the egg is expelled when the levels of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) suddenly increase. Ovarian malignancy, multiple births, ovarian hyperstimulation syndrome (OHSS), and premature delivery are associated with the usage of fertility medicines [13]. In this account, laparoscopy, hysteroscopy, and fallopian tube operations are considered for restoring female fertility via surgery. IUI and IVF are alternative ARTs for women. Medication therapy, surgery, hormone treatment, and ART are considered for the treatment of male infertility [14]. Sperm can be removed carefully from the testis or epididymis in persons with azoospermia. Accordingly, the varicocele can also frequently be treated with surgery. Furthermore, in certain circumstances, male reproductive problems cannot be cured, and men are unable to father children unless they donate their sperm or give it up for adoption. Considering these limitations, infertility requires improved potential solutions.

3 Stem cells express a novel goal in cell treatment

Recent advancements in the in vitro propagation of male germ cells from pluripotent stem cells has been considered, in which stem cells can be employed for ovarian regeneration and oocyte production in female infertility [15]. Stem cells are undifferentiated cells that occur in fetuses, adults, and embryos and are responsible for the production of differentiated cells. They are usually composed of adult tissues and early embryogenesis cells. During the post-natal and adult phases of development, tissue-specific stem cells are located in differentiated systems and play a crucial role in organ regeneration. Embryonic stem cells (ESCs), MSCs, spermatogonial stem cells (SSCs), and induced pluripotent stem cells (iPSCs) are the primary kinds of stem cells [16]. Totipotent cells are common undifferentiated cells, which can only be detected in the initial phases of growth. The fertilized egg and the first two dividing cells are totipotent, because they divide into extraembryonic and embryonic tissues that can generate embryos and the placenta. Correspondingly, iPSCs can differentiate into three embryonic stem cell lines, comprising endoderm, mesoderm, ectoderm, and from the mesoderm, tissues and organs grow [17].

Stem cell role has five main forms. The first sample entails the replacement and repair of damaged and dead cells. After being injected into the human body, stem cells self-aggregate in damaged organs and associated parts and develop into cell types that are local to these organs and parts. When SSCs are transplanted into sterile testes, their homing ability drives them to their niches. The transplanted SSCs subsequently attach to the Sertoli cells and form a tight connection with the blood-testicular barrier before migrating into their niche on the basement membrane [18]. Considerably, the activation of dormant and inhibiting cells is another kind. Cell division is the process by which the human body grows and develops. Some cells avoid undergoing regular cell cycles following division as they age and enter a condition of effective dormancy. Stem cells can stimulate dormant and suppressor cells, encouraging them to re-enter the cell cycle and divide to proliferate. This process increases the number of additional cells in the body and either normalizes or reverses the metabolic functions. Preclinical mice studies of chemotherapy-induced premature ovarian failure (POF) have indicated that these transplanted stem cells can reside in ovarian tissues and save the ovarian role. However, these pathways need to be examined further [19]. The third category includes the paracrine production of numerous enzymes, proteins, and cytokines to increase cellular proliferation, suppress efficient cell death, and differentiate existing tissue progenitor cells into tissue cells to repair injured tissue and produce new tissues. Spermatogenesis is a mechanism that is controlled by testosterone, endocrine, and paracrine secretion/autocrine elements such as the IL-1 family [20]. Pro-inflammatory cytokines are elevated, thus adversely affecting spermatogenesis. The development of the testicular paracrine/autocrine component and its control mechanism should be recommended for future male infertility therapeutic approaches [21]. The fourth kind entails the exertion of an immunosuppressive mechanism via cell–cell interaction and the production of soluble substances, thus inhibiting the growth of natural killer cells. The fifth kind includes encouraging intercellular signaling restoration. Impressively, the cell’s signal molecule combines with the receptor protein on the cell membrane, resulting in a complex formation in the receptor and the creation of a new signal substance within the cell. This process induces a response, along with an increase in ion permeability, a modification in cell shape, or a modification in another cell process [22].

4 Stem cells derived from diverse tissues

4.1 Induced pluripotent stem cells (iPSC)

Cell-based treatment is improved through the confirmation of pluripotent stem cells from fibroblast cell culture with the aid of different transcriptional regulators, such as Oct4, klf4, sox2, and c-myc. Through this process, the iPSCs produced through reprogramming resemble ESCs in terms of morphology, surface receptors expression, telomerase activity, the ability to differentiate into all three lineages, and the presence of a normal karyotype for development. Meaningly, iPSCs are superior to ESCs in regenerative medicine, because they are derived from adult cells, thus eliminating the ethical concerns associated with the use of embryos; these cells are commonly accessible. Considering that iPSCs are created from patients’ somatic cells, the risk of immunological rejection is low [23,24]. Various research has been undertaken to investigate the in vitro differentiation of iPSCs to male germ cells. Keratinocytes and cord blood were developed into haploid gamete-like cells. For the first three weeks, retinoic acid (RA) was employed in the culture media to establish differentiation. The cells were then extracted for another 2, 3, and 4 weeks during the administration of forskolin, recombinant leukemia inhibitory factor (LIF), and the CYP26 inhibitor R115866 [25]. Ramathal et al. [26] cultivated human skin cells from azoospermic men and fertile men during the addition of BMP4, BMP8, RA, and LIF before using them as xenotransplants into the testes of immune-deficient nude mice created by busulfan therapy. The results show that the iPSCs transplanted into seminiferous tubules developed into germ cell-like cells (GCLCs), while cells from outside tubules did not. Primordial germ cells (PGCs) results in the production of sperm and ova [26]. Irie et al. [27] found that SOX17 is the principal mechanism for human PGCs, although BLIMP1 was repressive based on the characterization. Fragile X male and female patient-derived iPSCs were used in the current research. Human iPSCs might differentiate into hPGCs in the absence of activin A, CHIRON, BMP4, SCF, EGF, and LIF [28]. They separate human PGCs from other cells by using the indicators found on PGCs including EpCAM and integrin 6 [28]. Furthermore, fibroblast-derived iPSCs can be differentiated into spermatogenic cells whether in a standard growth media or an in vivo xenotransplantation approach [29]. iPSC differentiation can be employed into oogonia, providing hope to couples with insufficient ovarian function or the inability to generate viable eggs [30]. In these experiments, somatic cells were first reprogrammed into iPSCs, and then incipient mesoderm-like cells (iMeLCs) were developed during the addition of activin A and Chiron. Afterward, human primordial GCLCs (hPGCLCs) were generated by plating iMeLCs in clumps. These hPGCLCs were cultivated for four months with gonadal cells obtained from female mice embryonic ovaries to produce oogonia through in vitro gametogenesis. The creation of oogonia from human pluripotent stem cells will usher in a new era in infertility treatment. Sugawa et al. [31] recorded the mechanistic explanations for human pluripotent stem cell differentiation to heterogeneous mesoderm-like cells in the form of cytokines accompanied by the transition of PGC-like cells (PGCLCs), and the PGCLCs are dedicated to improving gametes that are affiliated with a unique expression of PRDM14 [18,3134]. Despite repeated test findings, the teratogenic potential of iPSCs and their derivatives arising from reprogramming by inducing with tumorigenesis and the use of nucleic acid integration techniques limit their therapeutic use to customized cell-based treatment [35]. Furthermore, the persistence of epigenetic imprints and downregulation and the genomic instability may pose a remarkable barrier to their potential therapeutic effects [34,36]. Although the iPSC system does not damage embryos, the possibility of exploiting embryos produced from gametes advanced after iPSC reprogramming necessitates ethical approval from a review committee and the collection of explicit consent from the cells or tissue donation process of obtaining any sample for the advancement of iPSC for clinical purposes. Furthermore, IACUC permission is needed for their use in animal studies [37].

4.2 Mesenchymal stem cells

To adequately address the use of MSCs, the International Society for Cellular Therapy’s Mesenchymal and Tissue Stem Cell Committee set fundamental criteria for the characterization of human MSCs. MSCs need to be plastic-adherent whenever cultured in conventional circumstances. MSCs should exhibit CD105, CD73, and CD90 but not CD45, CD34, CD14 or CD11b, CD79a or CD19, or HLA-DR surface molecules. Finally, in vitro, MSCs have developed into adipocytes, chondroblasts, and osteoblasts [7].

Considerably, the large number of MSCs is grouped according to their source. MSCs are now being used in scientific research and clinical studies, either alone or in combination with other treatments, for possible applications to ovarian malfunction and endometrial problems [38,39]. Treatment agents for various disorders affecting female genitalia are generating a lot of interest. More fundamentally, these findings suggest a promising experimental method for exploring the underlying process of employing MSCs for the treatment of female infertility (Fig.2).

MSCs can be considered to migrate directly and impulsively to the wounded ovary and survive there under the encouragement of numerous stimuli, facilitating ovarian healing. The quantity of differentiated and functionally integrating MSCs is insufficient to account for the occasions in ovulation. Moreover, it is uncertain whether MSCs develop into oocytes after moving to damaged tissue. The increased ovarian efficiency in these trials could be attributed to paracrine processes. These methods entail the release of cytokines, such as vascular endothelial growth element, insulin-like growth factor, and hepatocyte growth factor, which aid in anti-inflammation, antiapoptotic, angiogenesis, antifibrosis, and immunoregulation, and consequently aid in ovarian repair. Conversely, MSCs increase endometrial reserve, which is primarily determined by homing and paracrine activities. Generally, the paracrine action of MSCs, instead of differentiation, is the most significant in research. The regenerative characteristics of transplanted MSCs can be linked to cell-to-cell contact and the production of bioactive chemicals that enhance tissue regeneration and angiogenesis, regulate inflammation, prevent fibrosis and immunological responses, and activate tissue-specific progenitor cells. MSCs engraft the endometrium in mice and humans. Subsequently, they differentiate into epithelial, stromal, and endothelial cells. Interestingly, MSCs may enhance endometrial regeneration and repair fertility through paracrine factors, although alternative methods are possible. Interestingly, different microRNAs and genes alongside their origination and performance are of great importance (Tab.2).

4.3 Embryonic stem cells (ESC)

ESCs perform a notable function in regenerative medicine. Human embryonic stem cells (hESC) are provided from the blastocyst inner cell mass and express the transcription portion Oct 4 [40]. Both mouse and human ESC increase into primordial germ cells in vitro, where they undergo meiosis and give birth to male and female gametes [41]. The feasibility of the development of relevant sperm from gene-corrected ESC extracted from cloned blastocysts that are derived from nuclear transferred somatic cells (ntESC) utilizing gene restoration methods has been demonstrated in an egg cell mouse form [42]. As a result, ESCs have emerged as an essential mechanism for cell-based treatment to address infertility concerns. After the first derivation, it has not been widely employed in cell replacement therapy because of ethical issues. Importantly, hESCs participate in endometrial destruction repair and use recovery [43].

4.4 Spermatogonial stem cell (SSC)

Through self-renewal and unrestricted differentiation into spermatogonia accompanied by haploid spermatozoa, SSCs serve as a critical role in regulating the highly efficient complicated procedure described as spermatogenesis in the seminiferous tubule [44]. Oocytes are fertilized by such differentiated spermatozoa. The strictly regulated spermatogenesis mechanism in mammals has been divided into three stages, namely, the proliferative phase (mortification of spermatogonia), the meiotic phase (recombination of genetic material), and spermiogenesis (transformation of spermatids into male gametes) [45]. Infertility can occur from any aberration in these regulated stages of spermatogenesis [46]. SSCs are formed from primitive germ cells that move to the gonadal ridge throughout fetal development and eventually lodge into the seminiferous tubules of mature testicles to aid in continuous sperm creation, according to a model system [47,48]. Although SSCs are an excellent tool for the treatment of infertility, they are not widely used during stem cell therapy because of their limited number in the testes and the challenging detection process and effective isolation and cultivation processes [17]. These SSCs might be separated and defined by technological advancements and their species-specific identifying indicators. For instance, in the separation of SSCs from mice [49], stage-specific embryonic antigen-1 (SSEA1), integrins 1 and 6 with rats [50], and SSE4 and G-protein coupled receptor 125 (GPR 125) from humans [51], spermatogonia-specific identifier Stra8 was used.

During spermatogenesis, SSCs can provide viable sperm, suggesting their effective use for the treatment of some instances of male infertility, particularly those caused by chemotherapy. Notably, autologous SSCs have been transplanted into 18 adults and 5 pre-pubertal non-human primates, who had been rendered sterile by chemotherapy. SSCs were found in the ejaculates of approximately 75% of adult animals. Subsequently, by using intracytoplasmic sperm injection, allogenic donated SSCs fertilized rhesus eggs, resulting in different phases of embryogenesis. This process had been an innovative achievement in the field of generative translational medication [52]. For the in vitro formation of possible stem cells into germ cells, various biomedical environments have been used, alone or in conjunction with anyone else, such as retinoic acid for correct timing in the onset of meiosis [53], CYP26 inhibitor for the regulation of meiosis across regulation of the STRA8 gene [54], testosterone for the possible release of stem cell component to stimulate germ cell differentiation [54], and forskolin for enhanced germ cell proliferation [55]. To confirm the potential of SSCs to establish colonies within the testes while also developing into male gametes via synchronized spermatogenesis, researchers have used both in vitro stimulation of stem cell proliferation transplantation into male mice that were made infertile [56]. The primary disadvantage of this cell-based treatment in fertility clinics is that it may disrupt the testes environment, lowering the acceptance of SSC transplantation and leading to therapeutic collapse [57].

4.5 Ovarian stem cells (OSC)

The discovery of the frequency of follicular atresia, the mortality of oocytes, and the degradation of ovarian function in mice led to the notion of OSCs. These actively reproducing cells were identified by Johnson et al. [58] depending on their ability to integrate 5-bromodeoxyuridine and the development of germ-cell detail related, MVH (mouse vasa homolog). Such mitotically active cells might promote follicle creation in the ovaries of mice in previous investigations [58]. A further study related FACs for the extraction of OSCs from the human ovarian cortex by using the stem-cell-specific marker VASA expression. Results show that after these cells were xenotransplanted into diabetic immunocompromised mice, they were able to improve follicle results [59]. The discovery of these cells defies the convention that only females are capable of prenatal folliculogenesis [60]. However, these cells have remained unidentified for a long time, owing to their small quantity, which accounts for 0.0145% of the total cell population in the mouse ovary, which falls with age. Furthermore, as contrasted to their male counterparts, these cells take a long time to differentiate in vitro [61]. According to recent work on the isolation of OSCs from elderly mice and subsequent folliculogenesis after transplantation in young mice [62], the use of OSCs may provide new hope to patients suffering from idiopathic fertility problems.

In age-related infertility and premature ovarian in pediatric trauma survivors during chemotherapy, OSCs restore fertility and give birth to live children [40,63].

5 MSCs were derived from various sources for the therapy

5.1 Endometrial stem cells (EndSCs)

Endometrial stem cells were first identified by Prianishnikov et al., through which the isolation of stem cells from endometrial tissues was then performed by Chan et al. [64,65]. Subsequently, based on other revealed indicators, they identified and cultivated MSCs from human endometrium and hypothesized the role in endometriosis and endometrial cancer [66]. The expression of CD44, CD73, and CD90 and the lack of CD34 and CD45 phenotypes distinguished stromal cells from epithelial progenitor cells, although SSEA1, N-cadherin, and NTDPase2 activity distinguished epithelial progenitor cells. Endothelial stem cells demonstrated the existence of a typical phenotype such as CD 31 and CD 34 [6769].

In the presence of inflammation or endometrial injury, these stem cells were guided to the wounded location via communication among chemokines and CXCR4, which is observed on stem cells, and CXCL12, which is provided by endometrial tissue epithelium [70]. Endometrial stem cells result in the formation of cells such as endometrial regenerative cells (ERCs), epithelial stem cells, MSCs, and endometrial side population cells (ESPs) that support endometrium renewal [71]. ESPs comprise 5% of EndSCs and are mostly seen during the menstrual cycle’s proliferative phase [72]. MSCs and other ERCs may differentiate into various tissues, including bones, AD, cartilage, and neural tissue [73]. In comparison with MSCs, they can effectively promote immunological modulation, angiogenesis, and the generation of matrix metalloproteinases. The preservation of several phases of reproduction, such as the growth of the endometrium, needs adequate blood circulation in the uterus [74]. In an animal transplantation system, poor neovascularization causes endometrial decreases, which is likely linked to the decreased discharge of vascular endothelial growth agents [75]. As a result, endometrial stem cells’ angiogenic properties might be implicated in reproductive medicine’s curative transplantation for endometrial regeneration, and menstrual blood can also be used to create endometrial stem cells (Fig.3).

Endometrial stem cells have been studied in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)- induced form of Parkinson’s disease [76] and animal models of insulin resistance [77] to see if they may restore the release of dopamine [67,71].

5.2 Umbilical cord MSCs (UC-MSCs)

CD29, CD44, CD73, CD90, and CD105 expression was found in Wharton’s jelly of umbilical-cord-derived MSCs, but CD31, CD45, and HLA-DR85 were not. Correspondingly [78], UC-MSCs have been widely used for cell-based treatment method for restoring fertility because of their low tumorigenicity, rapid self-renewal capability devoted to diverse mesodermal cell types, simple access to regularly-damaged resources, minimal ethical problems, and low immunogenicity [56]. Importantly, UC-MSCs repair ovarian reserve in multiple animal studies of early ovarian dysfunction by antiapoptotic action toward granulosa cells and hormone regulation, such as lowering FSH levels while increasing estrogen and progesterone rates [79,80]. By altering ovarian surface epithelium stability, inflecting the expression of cyto keratin 8/18, TGF-β, and proliferating cell nuclear antigen (PCNA), which are significant controls of follicular synthesis and the antiapoptotic activity of UC-MSCs, UC-MSCs could recover ovarian purpose in paclitaxel-induced ovarian dysfunction [81]. UC-MSCs help revive infertility in experimental rats of PCOS produced by dehydroepiandrosterone by inactivating proinflammatory cytokines such as IL-1, tumor necrosis factor-alpha (TNFα), and interferon-gamma (IFN-γ). The favorable effect of UC-MSCs on improving fertility could be associated with various signaling mechanisms. UC-MSCs stimulate the mitogen-activated protein kinase, G protein-coupled receptor, and insulin signaling pathways to inhibit granulosa cell death and increase fertility [82]. Endometrial cells can be formed from UC-MSCs [83]. UC-MSCs increase and decrease vascular and inflammatory markers, respectively, to repair fertility in an experimental animal of injured endometrium [84]. Conversely, in an experimentally induced uterine damage, UC-MSCs transplantation on a collagen scaffold might restore fertility by activating matrix metalloproteinase 9 [85]. The stimulation of primordial follicles via phosphorylation of transcription factors forkhead box protein O3a (FOXO3a) and FOXO1 by UC-MSCs on a collagen scaffold could improve ovarian treatment in patients with premature ovulatory failure (POF), as evidenced by an increase in follicle count [86]. Wharton’s jelly-derived MSC could increase the proliferation and reduce the apoptosis of mifepristone-induced destruction in endometrium obtained from women undergoing hysterectomy, according to Yang et al. [87], by upregulating VEGF expression and downregulating caspase 3 and 8 [87]. The effectiveness and safety of intramuscular injection of UC-MSCs have been widely studied for the therapies of uterine scars induced by cesarean section in a single-center, prospective, randomly selected double-blind, and placebo-controlled phase II clinical trial.

5.3 Amniotic fluid stem cells (AFSCs)

The amniotic fluid, which results in sustenance to the fetus, also provides a source of MSCs. The potential of AFSCs to differentiate into various cell types including adipocytes, osteocytes, and muscle, and their potential systemic immune capabilities make them ideal for regenerative medicine. Therefore, AFSCs have beneficial benefit in muscle and bone problems in various animal investigations [88,89]. AFSCs can help with infertility by restoring ovarian function through the activation of numerous signaling molecules, including VEGF, TGF, EGF, and morphogenic protein (BMP) [82]. AFSCs, particularly those with CD4C/CD105+ markers, inhibit follicular atresia, restore viable oocytes, and consequently preserve ovarian purpose in a mouse model of POF caused by chemotherapy [90,91].

5.4 Adipose-tissue-derived stem cells (ADMSC)

ADMSCs are a new form of MSC with biological features that are virtually identical to MSC produced from those other origins, but with greater benefits than BMSCs, since such tissues can be harvested with less invasive techniques and in bigger quantities [92]. As a result, they are frequently employed in reparative therapy for a wide range of clinical diseases. The effect of ADMSCs on ovarian grafts has been studied in a female animal model, and the results show that ADMSCs recovered ovarian use more quickly by increasing VEGF gene expression and encouraging angiogenesis in the transplanted tissue [93]. Across neovascularization and advancement of ovarian follicular proliferation, ADMSCs can recover ovarian function in an animal model of chemotherapy-induced ovarian destruction [94]. Inside an ovarian failure rat model, treatment of ADMSCs on a collagen scaffold resulted in improved ADMSC retention, resulting in a longer recovery of ovarian function and a higher conception rate relative to ADMSC injection [94]. ADMSCs, in mixture with hormonal treatment, succeeded in reducing fibrosis and induced endometrial restoration in an animal experiment of Asherman syndrome [95].

5.5 Amnion-derived MSCs (AmDMSCs)

In reproductive medicine, the amnion is a potential resource for cell-based treatment. Numerous investigations [96,97] have established the therapeutic effect of ADMSCs on an ovarian malfunction in chemotherapy-induced animal studies of POF. Considerably, AmDMSCs help preserve ovarian function by reducing apoptosis and enhancing granulosa cell proliferation and neovascularization in the external environment via paracrine processes. Furthermore, AmDMSC therapy works by inactivating the cytokines IL-1, IL-6, and TNF, which suppress inflammation. Human ADMSCs become much more effective in the recovery of ovarian function in a rat model of POF following therapy by using low-intensity pulsed ultrasound, according to Ling et al. [110].

5.6 Placenta-derived MSCs (PDMSCs)

PDMSCs have beneficial effects on folliculogenesis by modulating cytokines and different hormone production, including AMH, FSH, estradiol, and LH, and their receptors’ expression, according to several types of research done on an experimental animal of POF [84,98]. However, Li et al. [99] found that PDMSC alleviates ovarian insufficiency by involving the inositol-requiring enzyme 1 (IRE1) signaling pathway. PDMSCs are successful in 3D spheroid formation to achieve their restorative activity in ovariectomized rats, according to Kim et al. [100]. PDMSC differentiation may be influenced by the overexpression of miR222 [101].

5.7 Menstrual blood MSC (MB-MSC)

MSCs generated from menstrual blood (MB-MSCs) can multiply and differentiate into numerous lineages and self-renew. In addition, unlike other tissue-derived stem cells, the collection of these cells is non-invasive, safe, and simple, with few ethical concerns and minor immunological responses, making them suitable for clinical use in reproductive medicine. In an animal model, Liu et al. investigated the effects of MB-MSCs on cyclophosphamide-induced premature ovarian failure (POF). They discovered that when MB-MSC-treated rats were assessed to normal control, the number of regular ovarian follicles and the recovery of normal ovarian function increased as evidenced by the increased levels of ovarian hormones such as estradiol, antimullerian hormone, and inhibin. Researchers tested the busulfan-induced rat model of POF in the same way. They found that MB-MSC enhanced ovarian function by localizing MSCs into granulosa cells and increasing the expression of FSH receptors along with resting the ovary [102]. MSC generated from menstrual blood restored fertility in an animal model of injured endometrium by inducing angiogenesis and releasing anti-inflammatory mediators [103]. During the first period, Zheng et al. [104] demonstrated a theoretical foundation for the use of MB-MSCs in intrauterine adhesion therapy. In the laboratory, MB-MSCs could develop into endometrial cells, and the transplantation of these cells into NOD-SCID animals led to endometrium restoration. The inclusion of the OCT-4 transcription factor contributes to the differentiating potential of MB-MSCs. Zhang et al. [105] found that MB-MSCs combined with platelet-rich plasma could effectively restore fertility in mechanically injured experimental rats of intrauterine adhesion by significantly modulating the Hippo signaling pathway and its downstream regulators, including CTGF, Wnt5a, and Gdf5 [105].

Furthermore, by upregulating cyclin B1 and CDC2 protein levels and downregulating Gadd45b protein levels, MB-MSCs might ameliorate epirubicin (broad-spectrum anti-cancer drug)-induced ovarian destruction and thereby boost granulosa cell proliferation [106]. Tan et al. [107] noted on patients diagnosed with conferring resistance Asherman syndrome who were managed with MB-MSC transplantation accompanied by hormone treatment; five of these clinical cases resulted in an endometrial thickness of 7 mm, one of these 5 patients thought up spontaneously, and two of these seven patients construed since assisted reproduction innovation [106]. The downregulated expression of OCT-4 in MB-MSCs derived from individuals with severe intrauterine adhesion [104] supports the involvement of the OCT-4 transcription factor in MSC development found in animals [104].

5.8 Bone marrow MSCs (BMSC)

MSCs were isolated from bone marrow by using density gradient separation and then cultured in a growth medium for development since they were first documented in 1988 by Owen and Friedenstein [108]. In experimental rats, these BMSCs are devoted to the formation of endometrial and follicular cells in addition to mesodermal lineages [109,110].

Jing et al. used an experimental rat model to determine whether BMSCs may help in endometrial regeneration. They used rat tail intravenous injection to transplant BMSCs and observed a substantial increase in endometrial thickness and the expression of markers, including integrin 3, cytokeratin, leukemia inhibitor factor, and vimentin. The upregulation of anti-inflammatory cytokines such as fibroblast growth factor (bFGF) and interleukin-6 (IL-6) and the downregulation of proinflammatory cytokines such as tumor necrosis factor (TNF) and interleukin-1 (IL-1) were likely responsible for BMSCs’ beneficial effect [111]. Wang et al. [112] later acknowledged the medical applications of BMSCs in endometrial healing in 2016. Intrauterine adhesion was generated by mechanical damage in an experimental model, and BMSCs were administered either by a tail vein or directly into the uterus. Despite the elevation of estrogen and progesterone receptor expression, BMSCs could successfully cure endometrial damage in both situations, as demonstrated by the increase in the size of endometrial glands and the reduction in the fibrotic area [112]. Chemotherapy for cancer patients kills cancer cells and causes the death of granulosa cells, which are responsible for supplying sustenance to eggs through the production of estrogen and other paracrine hormones, resulting in ovarian failure. In a rabbit model of cyclophosphamide-mediated ovarian dysfunction, Abd-Allah et al. reported the therapeutic use of BMSCs in 2013 and investigated the underlying principle. They found that intravenous injection of BMSCs restored the shape and role of ovarian follicles by lowering FSH and increasing estrogen and VEGF levels. In 2017, Badawy carried out a comparative research on a white mouse model and found that 21 days after BMSC treatment, hormone levels recovered, and conception was observed in infertile mice. In a potential, experimental pilot testing on patients with Asherman syndrome/endometrial atrophy, Santamaria et al. [113] found that injecting CD133+ BMSCs into the spiral arterioles of recruitment of new patients improved endometrial thickness, the intensity of matured blood vessels, and the nature of infertile patients’ menstrual period. Furthermore, 10 of the 16 patients conceived naturally or after embryo transfer [113]. Therefore, BMSCs may give fresh chance for biological offspring in situations of ovarian or uterine failure.

6 Application of stem cell therapy in disease therapy

6.1 Azoospermia

Azoospermia, or the lack of mature, morphologically normal, and functioning sperm in the ejaculate, is responsible for approximately 15% of infertility caused exclusively by male causes [114]. Obstructive azoospermia (OA) and non-obstructive azoospermia (NOA) can be distinguished based on the patient’s medical history, genetic analysis, extensive thorough physical, and hormone assay. Surgical treatment can be used for the treatment of OA. However, NOA results in testicular failure, which could be caused by pathology primarily in the testis or secondary to the decreased release of gonadotropin from the pituitary. In the case of NOA, the success rate of ART by intracytoplasmic injection is very poor because of the difficulty in retrieving functional sperm. Stem cell therapy can provide the opportunity of having biologically-related offspring to NOA patients [115].

Considering the self-renewal capabilities of spermatogonial stem cells (SSCs), they can develop into haploid spermatids and then mature into spermatozoa, thus boosting spermatogenesis to address this commonality of male infertility [116]. During the first period, Brinster and Zimmerman [117] demonstrated that SSCs could improve fertility in a mouse model [117]. Later, Hermann et al. [52] revealed that SSCs injected into the rete testes of pre-pubertal rhesus monkeys grow into mature spermatozoa in tandem with the monkey’s maturity; the spermatozoa are detectable in the ejaculate and may fertilize [52]. However, considering the difficulties in identifying SSCs in the testes, the absence of an appropriate culture and maintenance technique for SSCs, and safety issues for patients after transplantation, SSC transplantation could not be repeated in people [115]. In addition to SSCs, embryonic and adult stem cells can develop into germ cells [24] and fertilize in laboratory animals [118,119]. Furthermore, in the case of hESCs, ethical issues and the teratogenicity of iPSCs limit their medicinal utility [24].

Several clinical studies involving the injection of bone-marrow-derived MSCs into the rete testis of azoospermia patients have been conducted or are now underway to measure the levels of hormones, testicular size, and sexual potency.

6.2 Polycystic ovarian syndrome (PCOS)

Rotterdam used two criteria to diagnose PCOS, which is a gynecological illness of reproductive-aged women caused by the following hormonal imbalance, presence of high levels of androgens, and ovulatory failure, or polycystic ovaries. Abnormal hair development on the body, irregular menstruation, infertility/subfertility, and obesity are all symptoms of PCOS [120]. Correspondingly, miR-320 regulates ovarian steroidogenesis by targeting E2F1 and SF-1 [98]. For the recovery of regular menstruation and fertility, hormonal contraceptives and clomiphene citrate are the first-line treatments [120]. The prospective involvement of stem cells for the treatment of PCOS symptoms has been established in various animal settings. MSCs suppress inflammation via the production of anti-inflammatory cytokines, which attenuate DHEA-mediated PCOS in mouse [78] and human theca cells from PCOS patients [121]. In a drug-induced mouse model of PCOS, Igboeli and colleagues [122] demonstrated that hMSCs may reverse and recommence the estrus cycle.

6.3 Endometriosis

Endometriosis is an estrogen-dependent complicated illness that affects pregnant women. Accordingly, approximately 10% of females have endometriosis, and roughly 50% of those who have endometriosis have difficulty conceiving [123]. The fertility rate in couples with endometriosis is much lower than that in normal fertile couples based on the Practice Committee of the American Society for Reproductive Medicine [124]. Retrograde menstruation, metastatic spread, coelomic metaplasia modified stem cells, immunology, and genetics are among the leading theories for endometriosis etiology [125]. Endometriosis, with its inflammatory response and elevated cytokine release, may influence fertility by altering ovum formation, gamete transit via tubal obstruction, and endometrial growth via regulation of the Wnt pathway [126].

Remarkably, the treatment of infertile couples with endometriosis with a GnRH agonist accompanied by IVF with or without ICSI improved pregnancy rates considerably [127]. However, later research by Benschop et al. [128] observed no clear improvement in infertility when hormone treatment was first administered. Surgical treatment alone or in combination with medical treatment showed different responses. Current scientific findings in both human and animal models have shown that the use of stem-cell-based treatment for the restoration of fertility in endometriosis patients can be effective [125].

6.4 Premature ovarian insufficiency (POI)

POI is a medical and biological factor that contributes to infertility. POI is diagnosed by “oligo/amenorrhea for at least 4 months and an increased FSH level > 25 IU/L on two occasions > 4 weeks apart,” based on the European Society of Human Reproduction and Embryology (ESHRE) standards [129]. POI affects approximately 1% of women of reproductive age (under 40 years), whereas the frequency is roughly 0.1% in women below the age of 30 years and 0.001% in women below the age of 20 years. Hypoestrogenism can have varying degrees of effect on the affected person’s physical and mental health. Nonetheless, in some circumstances, cryopreservation of oocytes before gonadotoxic therapy has been used to restore fertility. Ovarian vitrification is accompanied by in vitro activation by stimulation of the AKT pathway, followed by re-transplantation, which can retain fertility in patients with POI [130]. Furthermore, the effective demonstration of ovarian stem cells in both preclinical animal and human models may usher in a change in the possible therapy for infertility repair in POI patients [58]. Therefore, MSC stem cell therapy can be used in female reproductive problems (Tab.3).

6.5 Asherman syndrome (AS)

In 1950, Joseph Asherman was the first to report a gynecologic condition, in which a women had amenorrhea after a uterine cavity injury. Infertility, repeat abortion, and persistent pelvic discomfort with amenorrhea are all symptoms of Asherman syndrome (AS), which is defined by intrauterine adhesion and partial or full obliteration of the uterine cavity [131]. AS is caused by postpartum curettage in 90% of patients [132]. Three-dimensional ultrasonography, magnetic resonance imaging, hysterosalpingography, and hysteroscopy are some diagnostic options for this condition, and hysteroscopy is the gold standard. Preservation of the uterine cavity with hysteroscopic adhesiolysis and replacement of normal endometrial lining with hormonal treatment are two therapies for infertility caused by AS [133,134]. The surgical excision of AS has already been linked to several obstetric problems, including premature birth [135].

Consequently, the use of stem cell therapy for the treatment of AS has been investigated. In animal models [95] and clinical investigations [151], the use of stem cells obtained from bone marrow [136], autologous menstrual blood [107], and MSCs [137] for endometrial replenishment has been studied. In an animal model of injured endometrium, endometrial MSCs generated from menstrual blood increased fertility [103]. After transplantation of menstrual-blood-derived stem cells, women with AS were able to achieve regular menstruation [138] and endometrial regeneration [107].

7 Therapeutic performance of MSC-derived extracellular vesicles in reproductive diseases therapy

Adult stem cells called MSCs can be extracted from various tissues, including endometrial tissue, bone marrow, AD, menstrual blood, and UC [139]. MSCs have emerged as promising candidates for cell therapy in regenerative medicine because of their propensity for self-renewal and differentiation [140,141]. MSCs are effective in a range of illnesses, including diabetes, female reproductive abnormalities, cardiovascular disorders, renal fibrosis, and neurological diseases [142,143], in both preclinical and clinical investigations. Nevertheless, stem cell treatment may cause several drawbacks, such as inconvenient transportation or storage, marketing challenges, transplant rejection, and safety concerns if necessary monitoring tests are not performed [144,145]. EVs are lipid bilayer particles that are released from cells into the milieu and act as messengers by transporting various payloads, including microRNAs, proteins, cytokines, and lipids [146,147]. In comparison with MSCs, MSC generated extracellular vesicles (MSC-EVs) have identical functionalities to the parent cells and have greater biological stability and reduced immunogenicity [148,149]. MSC-EVs have great prospects in the reproductive treatment of different disorders and diseases, including IUA, PCOS, and POI. Remarkably, the therapeutic performances comprise immunomodulation, pro-angiogenesis, anti-oxidative stress, and anti-fibrosis. Accordingly, many studies have acknowledged the efficacy of MSC-EVs in developing female fertility for in vitro and in vivo cases, like influences before the utilization of MSC-EVs clinically [149].

8 Extracellular vesicles from MSC

Similar to almost all other cell types, MSCs may produce EVs [150]. Exosomes (50–150 nm), apoptotic bodies (500–5000 nm), and microvesicles (100–1000 nm) are the three kinds of EVs [151]. These subtypes have slightly different biogenesis mechanisms. Overall, endocytosis and exocytosis report for the biogenesis of most exosomes, initial endosomes can be established in endocytosis of plasma membrane, late endosomes can develop into multivesicular bodies (MVBs) filled with intraluminal vesicles (ILVs) after inward budding of endosomal membrane, and MVBs discharge ILVs upon fusion with the cell membrane, which is influenced by endo [152,153]. The production of MVs is very straightforward; it involves direct budding from the cellular membranes [154]. Exosomes originate from cell membrane protrusions [152,153]. As a result, more research into the process of EV biogenesis is required. Exosomes and MVs have different biogenesis, indicating that their chemical contents and activities also differ [155]. Phinney et al. suggested that mitochondria may be delivered to macrophages via MVs rather than exosomes to improve mitochondrial bioenergetics [155]. Consequently, in terms of the therapy of female reproductive problems, the biological components of exosomes and MVs could alter these merits (Fig.4).

9 Therapeutic effects of MSC-EVS in reproductive diseases

9.1 POI

POI is described as ovarian senescence in women under the age of 40 years, which is defined by amenorrhea or oligomenorrhea for at least 4 months, an increased FSH level (25 IU/L), and fluctuant estradiol (E2) [156,129]. It affects approximately 1% of women under the age of 40 years, resulting in poor infertility results and putting a strain on young couples [156,157]. Aberrant immunity, genetic abnormalities, environmental toxins, and chemotherapy or radiation may cause POI [158,159]. Considering that primordial follicles cannot be produced and the ovarian reservoir is defined at birth, ovarian function can be readily jeopardized by fast activation of primordial follicles, ovarian follicle depletion in the resting pool, and aberrant follicular atresia [160].

The X chromosome and several genes are required for follicle formation and expansion [161,162]. Inactivation of the SMC1B, or REC8 genes, STAG3 involved in meiosis causes oocyte arrest with POI [163,164]. Accordingly, Jaillard et al. presented novel candidate POI genes such as NRIP1, XPO1, and MACF1 via whole exome sequencing (WES) study [165]. A single causal component is hard to determine, because numerous genes interact in complex ways during folliculogenesis [160]. Aside from gene mutation, aberrant epigenetic alteration may also participate in atresia follicle growth [166]. As a result, abnormal gene expression or control in women can disrupt folliculogenesis and cause follicular atresia, which might progress to POI.

Signaling pathways including the TGF-β, PI3K/AKT/mTOR, and Hippo pathways play important roles in follicular growth [167,168]. Grosbois et al. discovered the synergistic effect of the PI3K/AKT and Hippo signaling pathways, in which primordial follicle recruitment was increased, resulting in fast follicle exhaustion [168]. The overexpression of AKT/mTOR could be induced by abnormal PI3K pathway activation, whereas the dephosphorylation of YAP/TAZ could be induced by disrupting the Hippo pathway; both processes lead to huge and premature development of primordial follicles [168]. The suppression of the PI3K/AKT pathway by depleting protein kinase Ck2 resulted in large follicular atresia in mice ovaries, demonstrating the need for a balanced PI3K/AKT system. Follicular atresia was also increased by an excessive autoimmune or inflammatory response [156,169]. Follicular atresia, for example, arises when GCs are continuously exposed to proinflammatory cytokines such as IFN-γ [170]. Additional conditions, such as an infinite quantity of certain chemotherapeutics, environmental contaminants, and reactive oxygen species (ROS), may also contribute to follicular atresia or depletion [159,160,171]. The actual cause of decreased ovarian function is unknown, and the convoluted pathological process of the disease makes it difficult to cure. In POI animal models, MSC-EVs can salvage GC viability, decrease ROS levels, and repair follicular count [172175]. AFMSC-EVs rescue ovarian follicles from chemotherapy-induced gonadotoxicity. In chemotherapeutics-treated mice, miR-10a, a significantly abundant miRNA in AFMSC-EVs, increased the resistance to GC apoptosis or follicular atresia [176]. Sun et al. then discovered that UCMSC-EVs decrease cisplatin-induced GC apoptosis in vitro [177]. Furthermore, in a mouse study, UCMSC-MVs-treated groups had greater levels of AKT, P-AKT, VEGF, and IGF than non-treated POI groups, indicating that UCMSCMVs may stimulate angiogenesis and activate the PI3K/AKT signaling pathway in the ovaries [174]. Accordingly, PTEN, which negatively regulates the PI3K/AKT pathway, was downregulated in POI rat models following injection of BMSC-EVs or AFMSC-EVs [178,179], indicating that MSC-EVs may reduce GC apoptosis via the PTEN-PI3K pathway. Furthermore, MSCEVs decreased SIRT families and P53 to prevent GC apoptosis, which is triggered by cisplatin or cyclophosphamide (CTX) [86,180,181]. Huang et al. discovered that ADSC-Exo controlled the SMAD signaling pathway and saved GCs from apoptosis via mRNA and protein assays [172].

MSC-EVs also enhanced the outcomes of offspring in POI models [182]. In this study, Liu et al. discovered that POI mice in the UCMSC-EV transplantation group had better fertility, shorter time to pregnancy, and more offspring than the POI mice. Furthermore, the Y-maze test and new object identification task revealed that their kids had substantially identical cognitive characteristics. MSCEVs boost the fertility of POI mice without adversely affecting their offspring’s cognitive functioning [182].

Likewise, the therapeutic use of MSC-EVs in POI women is still in its early stages. The mechanism in which MSC-therapeutic EV affects POI remains unknown, thus requiring additional investigation. The therapeutic properties of MSC-EVs could not be fully extrapolated to human patients based on animal models with abdomen injections. A medical experiment recently indicated that MSCs based on collagen scaffold (CS) were transplanted into the ovaries of POI patients via a retrograde injection approach, implying that CS/MSC-EVs might also be transmitted via intra-ovarian injection [86]. However, the method’s safety and effectiveness have not been determined. Remarkably, when murine embryos were exposed to endometrial MSCs (endMSC)-EVs, Blazquez et al. found high blastomere count and hatching rate [183,184]. EndMSC-EVs increase the success of in vitro fertilization in an elderly mouse model [185]. In POI patients, IVF-embryo transfer (IVF-ET) is commonly used for supported conception [172]. Although the safety of children must be investigated, the combination of IVF-ET with MSC-EVs might be a novel strategy for assisting POI patients in conceiving.

9.2 PCOS

PCOS is a prevalent reproductive endocrine illness marked by hyperandrogenism, ovulatory failure, polycystic ovarian morphology, overweight, and insulin levels that affect 5%–20% of reproductive-age women [183,186]. The core of PCOS is hyperandrogenism [187,188]. Androgen abundance stimulates tiny antral follicle development and early luteinization, inhibiting dominant follicle selection and thus impairing ovulation [187,188]. For women with PCOS, anovulatory infertility is a severe difficulty, and support and facilitating procedures are considered the last choice for conceiving [189]. In vitro maturation procedure focused on heterologous follicular fluid and GCs supernatant (HFF/GC-IVM protocol) was recently discovered to increase the maturity rate of immature denuded oocytes, fertilization rate, and hatched blastocyst level [190]. Conversely, ADSC-EVs prevent apoptosis and boost proliferation in PCOS patients’ cumulus cells (CCs), whereas increased expression of miR-323-3p plays a role in exosomes [191]. Even so, a few cases of MSC-EVs are being used for PCOS treatment.

9.3 IUA

The destruction of the uterine basalis layer and subsequent obliteration of the endometrium by fibrous structures define IUA, often referred to as Asherman syndrome (AS) [192]. Reduced menstrual flow, repeated fetal death, abnormal placental implantation, and infertility are all symptoms of IUA [193]. Furthermore, the endometrium of individuals with repeated IUA is frequently thin, resulting in thin endometrial (TE) [194]. Chronic endometritis, retained placenta, artificial abortion, and curettage are possible causes of this syndrome, and they disrupt the uterine environment’s balance [195]. Endometrial fibrosis participates in the onset and development of IUA [196]. The damaged endometrium cannot be healed naturally, which can cause immunological activation and inflammation and an overabundance of extracellular matrix (ECM) protein [196,197]. As a result, chronic inflammatory irritation encourages the creation of abnormal avascular fibrotic regions, resulting in tissue hypoxia and obstructing endometrial healing [197].

The amplification of the transforming growth factor-β1 (TGF-β1)/Smad3 signaling pathway has been implicated in the development of IUA in several investigations [198]. Considerably, NF-κB, an inflammatory marker, has been considered a risk factor for IUA [199]. TGF-β and connective tissue growth factor-2 (CTGF/CCN2) expression levels were linked positively to NF-κB pathway activity in the endometrium of IUA subjects, according to Xue et al., and TGF-β transcription was reduced after suppressing the NF-κB signaling system [200]. Importantly, an abnormally active Wnt/b-catenin pathway stimulates TGF-β-mediated fibrosis and mediates fibrogenesis in the endometrium [201,202]. FOXF2 protein enhances fibrogenesis in the IUA by activating the Wnt/β-catenin pathway and upregulating collagen type V alpha 2 (COL5A2) transcription in the endometrium [203,204]. As a result, the interaction of several proteins or signaling pathways plays an important role in the pathogenesis of IUA.

Fibroblasts, stromal cells, mast cells, smooth muscle cells, endothelial cells, M1 macrophages, and T cells were identified in the bioinformatic investigation of the uterus [205]. Identifying intercellular connections and cellular transdifferentiation might help researchers truly understand the pathophysiology of IUA and identify new therapy options. For example, a heatmap in this study revealed a significant level of contact between endothelial cells and fibroblasts or stromal cells, indicating that communication may be crucial in the evolution of IUA. Notably, the epithelial-to-mesenchymal transition (EMT) is critical for the evolution of IUA [201]. NUS1 protein overexpression increases EMT by modulating the AKT/NF-κB pathway, which may be inhibited by microRNA (miR)-466, according to research by using IUA rat models [206]. Moreover, miR-1291 increases endometrial fibrosis by inhibiting ArhGAP29 and upregulating the RhoA/ROCK1 pathway, and this process is also involved in EMT [207]. Furthermore, the Hippo/TAZ signaling pathway adversely influences the EMT process to some extend [201]. In this case, the activation of the Hippo pathway results in phosphorylated TAZ, thus inhibiting the EMT process [201]. Zhu et al. also discovered that the Hippo system, which would be activated by menstrual blood-derived stem cells (MenSCs), would block TGF-β-mediated stimulation of endometrial stromal cells (ESCs) and myofibroblast phenotypes [208]. Overall, AS needs to be treated by managing the phenotypic alternation in the endometrium, which is implicated in the pathological changes of IUA.

In animal and in vitro research, the effects of MSC-EVs on AS have been studied [209211]. In rat IUA models, Yao et al. discovered that BMSC-EVs increase endometrial glands, reduce fibrotic area, and even reverse EMT [212]. The injection of BMSCs-EVs decrease vimentin (VIM) levels while increasing cytokeratin (CK) 19 levels. Similarly, TGF-β1, TGF-β1R, and Smad2 expression were reduced in the intervention group, demonstrating that BMSC-EVs would heal endometrium by suppressing the TGF-β1/Smad2 signaling pathway [122]. Saribas et al. found that injecting uterus-derived MSC (uMSC)–EVs into the uterine cavity increased angiogenesis in IUA rats by boosting the expression of vascular markers CD31 and VEGFR1 [213]. UCMSC-EVs boosted rat TE restoration by overexpressing VEGF, Bcl-2 levels, and lowering fibrosis area, accordingly, indicating that MSCEVs might improve endometrial regeneration [214]. MSC-EVs may also stymie IUA progression by suppressing inflammation. UCMSC-EVs suppress the inflammatory factors interleukin (IL)-1b, IL-6, and TNF-α while increasing the anti-inflammatory factor IL-10 [210]. The above research examined the implantation and conception outcomes to fertility reserve. The findings demonstrate that UCMSC-EVs might enhance these two numbers in IUA rats, implying that UCMSC-EVs could be useful in resolving infertility in patients with IUA [210]. Therefore, MSCEVs can be used for the treatment of AS by improving endometrial health, slowing the fibrosis stage, increasing angiogenesis, and acting as an immunomodulator. Generally, the feature of stem cells as a main therapeutic category in infertility is indicated in Tab.4.

10 Role of personalized medicine in reproductive diseases

Personalized medicine was initially coined in the field of genetics more than a decade ago, and it is a desired therapeutic outcome of human genome sequencing. Personalized medicine relates to the capability to design medication based on molecular information to analyze disease patterns and diagnose disease. It also aims to effectively tune prevention and treatment interventions with less side effects. Personalized medicine has numerous definitions in this context. Precision medicine, which is described as the classification of patients with a specific disease into subpopulations that differ in their response to a given treatment based on phenotypic evidence, including biomarkers or genomes, is sometimes used interchangeably with this phrase. Personalized medicine can apply to the management of individual patients depending on various contextual elements, including therapeutic response, patient preferences, and established phenotypic results. The regulatory process for the commercialization of a novel treatment or a novel purpose for an existing drug necessitates a favorable benefit-risk profile and strong proof of efficacy. The purpose is based on the regulatory approval’s eligibility criteria and findings of the clinical trial. In comparison with personalized medicine, which may be focused on a single patient, precision medicine medications are frequently produced with diagnostic techniques that are required for the identification of a subgroup of patients [215,216].

Relatively, this new technology has always been a criterion in every operation in reproductive medicine, involving special analysis, identifying healthy embryos, correct implantation, and etiology-oriented evaluation. An uncharted zone of reproductive medicine will be explored, and the processes underlying the unknown causes of infertility will be described using genetic information and a massive amount of biological data. Precision medicine has become a blueprint for medical advancement, particularly in the field of reproductive health. In many other fields, the increasing availability of clinical and genetic biomarkers has aided clinicians in formulating overall illness predictions and making well-informed treatment decisions. For the treatment of chronic disease, a 4P (predictive, participatory, personalized, and defensive) health care approach has been proposed and is becoming increasingly popular. The expression of HER2 and ER can prognosticate the fate of a patient with breast cancer patient. Prognosis mainly aims to minimize the buildup of stresses and recognize early signs. Existing indicators in infertility treatment such as hormone levels and ultrasound scanning are still at the heart of illness prediction. The next generation of diagnostics, in conjunction with machine learning, will improve the precision with which dysfunctions are identified. The proteome, epigenetic organizations, genome, molecular prognosis, metabolome, and transcriptome can all be merged, and screening and recognition platforms can be built based on this data. The key to protection is to avoid infertility. Some of the reasons for infertility are linked to a sedentary lifestyle. Infertility can be prevented by increasing information about female reproductive health. With a better knowledge of the many reasons for infertility, more customized diagnoses and more accurate therapy can be provided, thus increasing the rate of success of ART. The patient psychological therapy and education need to be improved to solve emotional barriers and avoid dropouts that limit the understanding. Individualization of therapy, preventative treatments, and enhanced data gathering through self-tracking are important. These interventions can be realized by first obtaining as much knowledge of a person’s “health literacy.” Reproductive medicine will reach its peak as precision medicine continues to be promoted, offering benefits to more patients experiencing infertility [216,217].

11 Diagnostic genetic tests in reproductive disease

A succession of significant improvements in reproduction and scientific medicine has effectively linked these two professions in the previous few decades. From diagnostic techniques to the selection of the most sophisticated treatment, scientific medicine is essential in all stages of the evolution system.

In reproductive medicine, genetic testing is used for identifying the reasons for infertility, identifying genetic illnesses that are transmissible to kids, and optimizing assisted reproductive technologies.

Accordingly, infertile couples’ testing timelines now comprise biochemical and instrumental studies that permit for a prognosis in 65% of instances; genetic testing is used in the remaining 35% of unidentified cases. Considering that approximately 15% of genetic conditions are linked to infertility, and related clinical symptoms can have both genetic and non-genetic causative factors, an infertility prognosis should be made by using a combination of thorough health information and instrument and laboratory setting assessments, including aimed genetic screening [218]. Genetic assessment can help confirm the diagnostic accuracy, allowing for more specific and focused medical treatment.

12 Male genetic infertility

All three etiological classes of male fertility (pre-testicular, testicular, and post-testicular) have been linked to genetic variables. Conspicuously, upwards of 200 genetic diseases are associated with male infertility, ranging from the most prevalent clinical manifestations of infertility to the most complicated syndromes with signs and symptoms that go beyond reproductive issues [219]. Infertility is a clinical indication of a complicated disease; nevertheless, in certain hereditary disorders, infertility is the most prominent phenotypic trait. Furthermore, such infertile individuals need to be tracked over time, because they have higher mortality and worse life expectancy than the general public mainly because of the genetic defects that induce male sterility [220].

The existence of abnormalities in the stereogram is now the primary indication for genetic testing, especially in cases of severe oligospermia [221]. The number of genes indisputably connected to the more frequent phenotypes of oligozoospermia or azoospermia remains restricted (50%); the other half are genes implicated in teratozoospermia, despite the monomorphic variants of teratozoospermia being exceedingly rare [222].

Full chromosomal abnormalities (structural or numerical), partial chromosomal aberrations, and monogenic illnesses are all genetic disorders that are linked to male infertility. Importantly, sex chromosomal defects have a strong influence on spermatogenesis, whereas autosomal mutations are associated with teratozoospermia, or hypogonadism, and familial types of obstructive azoospermia.

The karyotype, the research of chromosome Y microdeletions, and the examination of the CFTR gene are the most popular genetic tests used for the diagnosis of male infertility. Although various mutations have been linked to male infertility, the underlying genetic etiology in 40% of all instances of male infertility is unclear [223]. Notably, the function of de novo mutations should be examined further, particularly in the wake of what occurs with Klinefelter syndrome and AZF alterations, which are virtually entirely caused by de novo mutations [222]. As a result, the entire diagnostic–therapeutic pathway of male infertility can be optimized and customized using genetic testing for the detection of particular clinical pictures, after suitable genetic testing: (1) for medical testing, (2) during medical decision to determine the most appropriate ART strategy.

13 Whole chromosomal aberrations

The frequency of chromosomal abnormalities ranges from 1.05% to 17% (depending on the study group) but is 0.84% in infants [224]. In terms of numerical abnormalities, structural chromosomal inversions are more prevalent; however, this does not relate to sex chromosomes, in which abnormalities, which account for approximately 4.2% of all chromosomal aberrations, are indicated by sex chromosome aneuploidies in 84% of the cases and structural rearrangements of chromosome Y in the remaining 16% of cases. The much more common kind of sex chromosomal aneuploidy discovered in infertile males is Klinefelter syndrome (karyotype 47, XXY); the second most common gonosomal abnormality is double Y syndrome or Jacobs syndrome, which is defined by the existence of the Y chromosome disomy [225,226]. In terms of providing a lower reproduction rate, people with chromosomal abnormalities are likely to have an abortion or have a kid with an abnormal karyotype.

14 Partial chromosomal aberrations

Microdeletions in the AZF (azoospermia factor) section of the Y chromosome (Yq) have indeed been discovered in 8%–12% of azoospermic males and 3%–7% of oligozoospermic males [222], leading to the most prevalent molecular gene responsible for male infertility [225]. Considering that the AZF area contains three sets of genes (AZFa, AZFb, and AZFc) that are involved in spermatogenesis, partial or full reductions in this domain may reduce reproductive potential. Interestingly, the sperm count (5106 spermatozoa/mL) linked with primary vestibulopathy indicates AZF deficiency detection, and ICSI is required to overcome infertility [227].

Considering that male children will inherit the same or worse Yq microdeletions as their fathers, genetic counseling is required [228]. Turner’s syndrome (45, X0) and other phenotypic malformations linked with sex chromosomal mosaicism [229] are risks that parents should be informed. Approximately 60% of all Yq microdeletions are caused by a reorganization of the AZFc zone [230]. The 3.5 Mb AZFc region includes multiple copies of five repetitions (b1, b2, b3, b4, and gr), whose familiarity and huge size predispose an individual to de novo deletions by homologous recombination [231]. The deletion of the entire b2/b4 area, which contains the DAZ family, is the most prevalent and causes spermatogenesis to deteriorate [231,232].

Although millions of genes are implicated in male infertility, only a few genetic illnesses (e.g., cystic fibrosis) have been studied [233]. The methods for identifying a single causal gene are ineffective considering that the testis just expresses over 2300 genes, many of which regulate reproductive activities and potentially contribute to male infertility. Single or several genetic abnormalities account for roughly half of all infertility cases, and 20% of patients have unidentified genetic reasons [218]. Additionally, considering that methods such as next-generation sequencing become more widely used for both diagnostic reasons, we will be able to quickly extend our understanding in this sector [234]. The clinical features and the primary genetic problems that may interfere with healthy reproduction need to be examined to enhance the focused genetic test in the presence of particular clinical images.

15 Female genetic infertility

Unlike male infertility, the genetic causes of female infertility are poorly understood. As a result, infertile females undergo fewer diagnostic tests to evaluate the existence of chromosomal diseases or single-gene problems linked to their clinical characteristics. Female infertility is more usually caused by syndromic disorders than by isolated infertility owing to hereditary factors. Genetic testing is now confined to chromosomal abnormalities and FMR1 premutation in patients with POI. Remarkably, more details have been reported about the primary chromosomal and genetic variations that could interact with healthy reproduction, and the primary phenotype extraction and laboratory tests that can be obtained in pre- and postnatal durations have been mentioned.

16 Whole chromosomal aberrations for female genetic infertility

Considering that chromosomal problems remarkably influence fertility and the chance of miscarriage, a karyotype study is typically recommended [235]. Robertsonian reciprocal translocations are abnormalities personally accountable for blocks of meiosis and structural modifications of the X chromosome, which are the most clinically meaningful structural abnormalities in infertile females. Infertility is considerably elevated in patients with reciprocal translocations, especially hypogonadotropic hypogonadism with primary or secondary amenorrhea or oligomenorrhea. The balanced conformational changes do not induce health concerns for their carriers, because they do not induce genetic information loss or duplication, but these conditions may result in gametes with imbalanced genetic data and possibly cause infertility or numerous miscarriages. Some chromosomal anomalies, including the XXX karyotype, are not strongly linked to infertility. Serious mistakes throughout crossing-over and/or meiotic nondisjunction in women with a normal karyotype generate a variable number of oocytes with chromosomal anomalies [236,237]. 45X, trisomy, and polyploidy are the three basic types of disorders. These occurrences increase with maternal age [224]. PGT allows for the analysis of gametes or embryos while undergoing ART. While checking for aneuploid embryos and exclusively transferring euploid embryos, the technique’s effectiveness is improved [238].

17 Fragile X syndrome

The existence of over 200 repeats of the CGG triplet pattern in the fragile X mental retardation 1 gene or a loss impacting the fragile X mental retardation 2 gene causes fragile X syndrome, which is an autosomal dominant genetic condition. Accordingly, menstrual instability reduces ovarian reserve, and early ovarian insufficiency is a symptom of female FMR1 premutation or FMR2 microdeletion [239,240]. In contrast to the family background, a molecular test should be discussed in the case of women with similar clinical signs. X-chromosome-linked abnormalities are the most prevalent genetic donors to POI. These conditions may be caused by a change in an autosomal chromosome [241]. Mutations should be identified immediately to manage reproductive choices and, if required, a preimplantation genetic diagnostic program should be selected to determine the particular clinical scenarios, in which a focused genetic test could help lead to a tailored diagnostic–treatment strategy.

18 Detection of hereditary illnesses that are transmissible to progeny by using molecular techniques

Perinatal mortality is caused by inherited chromosomal or genetic abnormalities in 20%–25% of instances [242]. Preconception carrier detection has been increasingly popular in recent decades as a result of increased medical understanding. The ability to identify couples at risk of passing on a certain hereditary illness to their children allows future parents to make educated reproductive decisions. If one of the reproductive partners has a gene mutation that causes one of the hereditary diseases, the pregnancy might result in a kid with that disease.

More than 2000 genetic abnormalities can be tested, including major disorders such as cystic fibrosis, spinal muscular atrophy, and sickle cell anemia and more complicated ailments such as mental retardation and congenital heart disease. In this setting, genetic counseling is critical for identifying genetic risk, correctly assigning patients, and alerting patients about genetic concerns that affect decision-making [243]. Preconception carrier examination provides genetic data in various illnesses, allowing all carrier couples to make reproductive options depending on their findings. The personalized genetic test is an important tool for women and their newborns to enhance short- and long-term results [244,245].

Therefore, in the involvement of carrier or afflicted spouses, various tests are available to determine a transmissible condition to the children during the prenatal period. Any one of these treatments can only be used during a certain point in pregnancy or at various stages of the IVF process.

Investigative PND is often conducted on DNA recovered from fetal cells acquired by chorionic villus sampling (CVS) or amniocytes, and the results are available in 7 to 15 days [246]. The molecular diagnosis for monogenic illness is carried out by direct mutation analysis when the parental mutations are recognized or by linkage analysis when the parental mutations are unclear [246,247]. In terms of the specified analytic steps, paternity verification and contamination analysis are always conducted [247].

The non-invasive prenatal diagnosis (NIPD) of monogenic illness, which can identify fetal genetic changes in maternal blood at an initial gestational age, has gained wide attention. NIPD remains a difficulty, although non-invasive prenatal testing (NIPT) by using cell-free fetal DNA (cfDNA) for the screening of chromosomes 21, 18, 13, X, and Y has been clinically used [248]. Under sex-related diseases and RHD, NIPD has just lately been adopted for therapeutic usage [249]. NIPD has been studied in several monogenic illnesses, including Duchenne, congenital adrenal hyperplasia, thalassemia, and Becker muscular dystrophy [250,251]. The prospect of adopting NIPD in clinical instances is being driven by the disruptive technology of NGS combined with the haplotyping technique.

Considerably, PGT has the same diagnosis and treatment motivation as traditional PND, with the added benefit of diagnosing at an earlier stage in the embryo. Curative abortion can be prevented by delivering only disease-free embryos to the mother. PGT appears to require the use of IVF methods, including the collection of gametes from both partners, fertilization of the oocyte by intracytoplasmic sperm injection (ICSI), embryo transfer, embryo biopsy that allows one or more cells from the blastomere or trophectoderm to be taken 3 or 5 days post-fertilization, and molecular analysis.

19 Conclusions

Stem cell treatment can now be used for the treatment of degenerative disorders, prevention of aggressive malignancies, and healing of damaged tissue. However, many areas of stem cell treatment are currently unclear, leaving the huge unrealized potential for use in managing disorders such as infertility. Based on the findings, once the correct technique is discovered, research will be suitable for the treatment of infertility. Ovarian-derived stem cells have a wide range of therapeutic applications. The therapeutic use of stem cells needs to be following ethical criteria, such as informed optional approval and other clinical investigation ethics guidelines. Whether a normal offspring can be created from gametes obtained from pluripotent stem cells is unknown. At the moment, stem cell-derived gametes can be utilized as an in vitro system to assess medication benefits. Generally, stem cell analysis has yielded significant new advances in the medicine of infertility. We shall keep on trying to disentangle the intricate web of ethical considerations surrounding this treatment. In certain circumstances, stem cell treatment may provide promise to people who would like to have their own genetically linked children. Embryonic stem cell transplantation has fallen out of favor because of ethical concerns and immunologic interference, pushing infertility doctors to look into additional stem cell treatments. Moreover, significant analysis is being done on induced pluripotent stem cells (iPSCs), which has few-to-no ethical problems and can produce person-specific haploid gametes. MSCs, which are produced from widely accessible resources such as AD, bone marrow, amniotic fluid, placenta, amnion, and menstrual blood and have few ethical problems, are gaining favor for use in fertility clinics. Several animal and human investigations have confirmed the importance of MB-MSCs in endometrial regeneration and ovarian performance recovery. Knowing the role of microRNAs in stem cell differentiation will provide a thorough understanding of stem cell modes of action for fertility restoration. Nonetheless, further large-scale clinical trials are required to demonstrate the safety and effectiveness of stem cell-based treatment in reproduction.

References

[1]

Hull MG, Glazener CM, Kelly NJ, Conway DI, Foster PA, Hinton RA, Coulson C, Lambert PA, Watt EM, Desai KM. Population study of causes, treatment, and outcome of infertility. Br Med J (Clin Res Ed) 1985; 291(6510): 1693–1697

[2]

Deroux A, Dumestre-Perard C, Dunand-Faure C, Bouillet L, Hoffmann P. Female infertility and serum auto-antibodies: a systematic review. Clin Rev Allergy Immunol 2017; 53(1): 78–86

[3]

Holmberg L, Iversen OE, Rudenstam CM, Hammar M, Kumpulainen E, Jaskiewicz J, Jassem J, Dobaczewska D, Fjosne HE, Peralta O, Arriagada R, Holmqvist M, Maenpaa J; HABITS Study Group. Increased risk of recurrence after hormone replacement therapy in breast cancer survivors. J Natl Cancer Inst 2008; 100(7): 475–482

[4]

Vermeulen RFM, Korse CM, Kenter GG, Brood-van Zanten MMA, Beurden MV. Safety of hormone replacement therapy following risk-reducing salpingo-oophorectomy: systematic review of literature and guidelines. Climacteric 2019; 22(4): 352–360

[5]

Practice Committee of American Society for Reproductive Medicine. Multiple gestation associated with infertility therapy: an American Society for Reproductive Medicine Practice Committee opinion. Fertil Steril 2012; 97(4): 825–834

[6]

Blum B, Benvenisty N. The tumorigenicity of human embryonic stem cells. Adv Cancer Res 2008; 100: 133–158

[7]

Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8(4): 315–317

[8]

Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells 2010; 28(3): 585–596

[9]

Körbling M, Anderlini P. Peripheral blood stem cell versus bone marrow allotransplantation: does the source of hematopoietic stem cells matter?. Blood 2001; 98(10): 2900–2908

[10]

In’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, Kanhai HH. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2004; 22(7): 1338–1345

[11]

Hansen KR, He AL, Styer AK, Wild RA, Butts S, Engmann L, Diamond MP, Legro RS, Coutifaris C, Alvero R, Robinson RD, Casson P, Christman GM, Huang H, Santoro N, Eisenberg E, Zhang H; Eunice Kennedy Shriver National Institute of Child Health, Human Development Reproductive Medicine Network. Predictors of pregnancy and live-birth in couples with unexplained infertility after ovarian stimulation-intrauterine insemination. Fertil Steril 2016; 105(6): 1575–1583.e2

[12]

Asemota OA, Klatsky P. Access to infertility care in the developing world: the family promotion gap. Semin Reprod Med 2015; 33(1): 17–22

[13]

Mourad S, Brown J, Farquhar C. Interventions for the prevention of OHSS in ART cycles: an overview of Cochrane reviews. Cochrane Database Syst Rev 2017; 1(1): CD012103

[14]

Kathrins M, Niederberger C. Diagnosis and treatment of infertility-related male hormonal dysfunction. Nat Rev Urol 2016; 13(6): 309–323

[15]

Smith RP, Lipshultz LI, Kovac JR. Stem cells, gene therapy, and advanced medical management hold promise in the treatment of male infertility. Asian J Androl 2016; 18(3): 364

[16]

Chen D, Gell JJ, Tao Y, Sosa E, Clark AT. Modeling human infertility with pluripotent stem cells. Stem Cell Res (Amst) 2017; 21: 187–192

[17]

Volarevic V, Bojic S, Nurkovic J, Volarevic A, Ljujic B, Arsenijevic N, Lako M, Stojkovic M. Stem cells as new agents for the treatment of infertility: current and future perspectives and challenges. Biomed Res Int 2014; 2014: 507234

[18]

Yoshinaga K, Nishikawa S, Ogawa M, Hayashi S, Kunisada T, Fujimoto T, Nishikawa S. Role of c-kit in mouse spermatogenesis: identification of spermatogonia as a specific site of c-kit expression and function. Development 1991; 113(2): 689–699

[19]

Kawamura K, Kawamura N, Hsueh AJ. Activation of dormant follicles: a new treatment for premature ovarian failure?. Curr Opin Obstet Gynecol 2016; 28(3): 217–222

[20]

Rozwadowska N, Fiszer D, Jedrzejczak P, Kosicki W, Kurpisz M. Interleukin-1 superfamily genes expression in normal or impaired human spermatogenesis. Genes Immun 2007; 8(2): 100–107

[21]

Huleihel M, Lunenfeld E. Regulation of spermatogenesis by paracrine/autocrine testicular factors. Asian J Androl 2004; 6(3): 259–268

[22]

Naylor CE, Bagnéris C, DeCaen PG, Sula A, Scaglione A, Clapham DE, Wallace BA. Molecular basis of ion permeability in a voltage-gated sodium channel. EMBO J 2016; 35(8): 820–830

[23]

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(2): 188–195

[24]

Hou J, Yang S, Yang H, Liu Y, Liu Y, Hai Y, Chen Z, Guo Y, Gong Y, Gao WQ, Li Z, He Z. Generation of male differentiated germ cells from various types of stem cells. Reproduction 2014; 147(6): R179–R188

[25]

Eguizabal C, Montserrat N, Vassena R, Barragan M, Garreta E, Garcia-Quevedo L, Vidal F, Giorgetti A, Veiga A, Izpisua Belmonte JC. Complete meiosis from human induced pluripotent stem cells. Stem Cells 2011; 29(8): 1186–1195

[26]

Ramathal C, Durruthy-Durruthy J, Sukhwani M, Arakaki JE, Turek PJ, Orwig KE, Reijo Pera RA. Fate of iPSCs derived from azoospermic and fertile men following xenotransplantation to murine seminiferous tubules. Cell Rep 2014; 7(4): 1284–1297

[27]

Irie N, Weinberger L, Tang WW, Kobayashi T, Viukov S, Manor YS, Dietmann S, Hanna JH, Surani MA. SOX17 is a critical specifier of human primordial germ cell fate. Cell 2015; 160(1–2): 253–268

[28]

Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, Yabuta Y, Kurimoto K, Ohta H, Moritoki Y, Iwatani C, Tsuchiya H, Nakamura S, Sekiguchi K, Sakuma T, Yamamoto T, Mori T, Woltjen K, Nakagawa M, Yamamoto T, Takahashi K, Yamanaka S, Saitou M. Robust in vitro induction of human germ cell fate from pluripotent stem cells. Cell Stem Cell 2015; 17(2): 178–194

[29]

Easley CA 4th, Phillips BT, McGuire MM, Barringer JM, Valli H, Hermann BP, Simerly CR, Rajkovic A, Miki T, Orwig KE, Schatten GP. Direct differentiation of human pluripotent stem cells into haploid spermatogenic cells. Cell Rep 2012; 2(3): 440–446

[30]

Gell JJ, Clark AT. Restoring fertility with human induced pluripotent stem cells: are we there yet?. Cell Stem Cell 2018; 23(6): 777–779

[31]

Sugawa F, Araúzo-Bravo MJ, Yoon J, Kim KP, Aramaki S, Wu G, Stehling M, Psathaki OE, Hübner K, Schöler HR. Human primordial germ cell commitment in vitro associates with a unique PRDM14 expression profile. EMBO J 2015; 34(8): 1009–1024

[32]

Sallam HN, Sadek SS, Agameya AF. Assisted hatching—a meta-analysis of randomized controlled trials. J Assist Reprod Genet 2003; 20(8): 332–342

[33]

Bharti D, Jang SJ, Lee SY, Lee SL, Rho GJ. In vitro generation of oocyte like cells and their in vivo efficacy: how far we have been succeeded. Cells 2020; 9(3): 557

[34]

Wu P, Deng G, Sai X, Guo H, Huang H, Zhu P. Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci Rep 2021; 41(6): BSR20200833

[35]

Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med 2013; 19(8): 998–1004

[36]

Fernandez Tde S, de Souza Fernandez C, Mencalha AL. Human induced pluripotent stem cells from basic research to potential clinical applications in cancer. Biomed Res Int 2013; 2013: 430290

[37]

Samadani AA, Keymoradzdeh A, Shams S, Soleymanpour A, Rashidy-Pour A, Hashemian H, Vahidi S, Norollahi SE. CAR T-cells profiling in carcinogenesis and tumorigenesis: an overview of CAR T-cells cancer therapy. Int Immunopharmacol 2021; 90: 107201

[38]

Naji A, Rouas-Freiss N, Durrbach A, Carosella ED, Sensébé L, Deschaseaux F. Concise review: combining human leukocyte antigen G and mesenchymal stem cells for immunosuppressant biotherapy. Stem Cells 2013; 31(11): 2296–2303

[39]

Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell 2015; 17(1): 11–22

[40]

Desai N, Rambhia P, Gishto A. Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reprod Biol Endocrinol 2015; 13(1): 9

[41]

Kehler J, Hübner K, Garrett S, Schöler HR. Generating oocytes and sperm from embryonic stem cells. Semin Reprod Med 2005; 23(3): 222–233

[42]

Yuan Y, Zhou Q, Wan H, Shen B, Wang X, Wang M, Feng C, Xie M, Gu T, Zhou T, Fu R, Huang X, Zhou Q, Sha J, Zhao XY. Generation of fertile offspring from Kit(w)/Kit(wv) mice through differentiation of gene corrected nuclear transfer embryonic stem cells. Cell Res 2015; 25(7): 851–863

[43]

Wang J, Liu C, Fujino M, Tong G, Zhang Q, Li XK, Yan H. Stem cells as a resource for treatment of infertility-related diseases. Curr Mol Med 2019; 19(8): 539–546

[44]

Kita K, Watanabe T, Ohsaka K, Hayashi H, Kubota Y, Nagashima Y, Aoki I, Taniguchi H, Noce T, Inoue K, Miki H, Ogonuki N, Tanaka H, Ogura A, Ogawa T. Production of functional spermatids from mouse germline stem cells in ectopically reconstituted seminiferous tubules. Biol Reprod 2007; 76(2): 211–217

[45]

Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod 2018; 99(1): 52–74

[46]

De Kretser DM, Baker HW. Infertility in men: recent advances and continuing controversies. J Clin Endocrinol Metab 1999; 84(10): 3443–3450

[47]

Kanatsu-Shinohara M, Lee J, Inoue K, Ogonuki N, Miki H, Toyokuni S, Ikawa M, Nakamura T, Ogura A, Shinohara T. Pluripotency of a single spermatogonial stem cell in mice. Biol Reprod 2008; 78(4): 681–687

[48]

McLean DJ. Spermatogonial stem cell transplantation and testicular function. Cell Tissue Res 2005; 322(1): 21–31

[49]

Guan K, Nayernia K, Maier LS, Wagner S, Dressel R, Lee JH, Nolte J, Wolf F, Li M, Engel W, Hasenfuss G. Pluripotency of spermatogonial stem cells from adult mouse testis. Nature 2006; 440(7088): 1199–1203

[50]

Hamra FK, Schultz N, Chapman KM, Grellhesl DM, Cronkhite JT, Hammer RE, Garbers DL. Defining the spermatogonial stem cell. Dev Biol 2004; 269(2): 393–410

[51]

Izadyar F, Wong J, Maki C, Pacchiarotti J, Ramos T, Howerton K, Yuen C, Greilach S, Zhao HH, Chow M, Chow YC, Rao J, Barritt J, Bar-Chama N, Copperman A. Identification and characterization of repopulating spermatogonial stem cells from the adult human testis. Hum Reprod 2011; 26(6): 1296–1306

[52]

Hermann BP, Sukhwani M, Winkler F, Pascarella JN, Peters KA, Sheng Y, Valli H, Rodriguez M, Ezzelarab M, Dargo G, Peterson K, Masterson K, Ramsey C, Ward T, Lienesch M, Volk A, Cooper DK, Thomson AW, Kiss JE, Penedo MC, Schatten GP, Mitalipov S, Orwig KE. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell 2012; 11(5): 715–726

[53]

Koubova J, Menke DB, Zhou Q, Capel B, Griswold MD, Page DC. Retinoic acid regulates sex-specific timing of meiotic initiation in mice. Proc Natl Acad Sci USA 2006; 103(8): 2474–2479

[54]

Griswold MD. The central role of Sertoli cells in spermatogenesis. Semin Cell Dev Biol 1998; 9(4): 411–416

[55]

Byskov AG, Fenger M, Westergaard L, Andersen CY. Forskolin and the meiosis inducing substance synergistically initiate meiosis in fetal male germ cells. Mol Reprod Dev 1993; 34(1): 47–52

[56]

Zhu Y, Hu HL, Li P, Yang S, Zhang W, Ding H, Tian RH, Ning Y, Zhang LL, Guo XZ, Shi ZP, Li Z, He Z. Generation of male germ cells from induced pluripotent stem cells (iPS cells): an in vitro and in vivo study. Asian J Androl 2012; 14(4): 574–579

[57]

Easley CA 4th, Simerly CR, Schatten G. Stem cell therapeutic possibilities: future therapeutic options for male-factor and female-factor infertility?. Reprod Biomed Online 2013; 27(1): 75–80

[58]

Johnson J, Canning J, Kaneko T, Pru JK, Tilly JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature 2004; 428(6979): 145–150

[59]

White YA, Woods DC, Takai Y, Ishihara O, Seki H, Tilly JL. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat Med 2012; 18(3): 413–421

[60]

Dunlop CE, Telfer EE, Anderson RA. Ovarian stem cells—potential roles in infertility treatment and fertility preservation. Maturitas 2013; 76(3): 279–283

[61]

Oatley J, Hunt PA. Of mice and (wo)men: purified oogonial stem cells from mouse and human ovaries. Biol Reprod 2012; 86(6): 196

[62]

Niikura Y, Niikura T, Tilly JL. Aged mouse ovaries possess rare premeiotic germ cells that can generate oocytes following transplantation into a young host environment. Aging (Albany NY) 2009; 1(12): 971–978

[63]

Anderson RA, Wallace WHB. Fertility preservation in girls and young women. Clin Endocrinol (Oxf) 2011; 75(4): 409–419

[64]

Prianishnikov VA. On the concept of stem cell and a model of functional-morphological structure of the endometrium. Contraception 1978; 18(3): 213–223

[65]

Chan RW, Schwab KE, Gargett CE. Clonogenicity of human endometrial epithelial and stromal cells. Biol Reprod 2004; 70(6): 1738–1750

[66]

Gargett CE, Schwab KE, Zillwood RM, Nguyen HP, Wu D. Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biol Reprod 2009; 80(6): 1136–1145

[67]

de Miguel-Gómez L, López-Martínez S, Francés-Herrero E, Rodríguez-Eguren A, Pellicer A, Cervelló I. Stem cells and the endometrium: from the discovery of adult stem cells to pre-clinical models. Cells 2021; 10(3): 595

[68]

Akbar Samadani A, Keymoradzdeh A, Shams S, Soleymanpour A, Elham Norollahi S, Vahidi S, Rashidy-Pour A, Ashraf A, Mirzajani E, Khanaki K, Rahbar Taramsari M, Samimian S, Najafzadeh A. Mechanisms of cancer stem cell therapy. Clin Chim Acta 2020; 510: 581–592

[69]

Rahimi S, Roushandeh AM, Ebrahimi A, Samadani AA, Kuwahara Y, Roudkenar MH. CRISPR/Cas9-mediated knockout of Lcn2 effectively enhanced CDDP-induced apoptosis and reduced cell migration capacity of PC3 cells. Life Sci 2019; 231: 116586

[70]

Lapidot T, Petit I. Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp Hematol 2002; 30(9): 973–981

[71]

Azizi R, Aghebati-Maleki L, Nouri M, Marofi F, Negargar S, Yousefi M. Stem cell therapy in Asherman syndrome and thin endometrium: stem cell-based therapy. Biomed Pharmacother 2018; 102: 333–343

[72]

Kato K, Yoshimoto M, Kato K, Adachi S, Yamayoshi A, Arima T, Asanoma K, Kyo S, Nakahata T, Wake N. Characterization of side-population cells in human normal endometrium. Hum Reprod 2007; 22(5): 1214–1223

[73]

Meng X, Ichim TE, Zhong J, Rogers A, Yin Z, Jackson J, Wang H, Ge W, Bogin V, Chan KW, Thébaud B, Riordan NH. Endometrial regenerative cells: a novel stem cell population. J Transl Med 2007; 5(1): 57

[74]

Gargett CE, Rogers PA. Human endometrial angiogenesis. Reproduction 2001; 121(2): 181–186

[75]

Numao A, Hosono K, Suzuki T, Hayashi I, Uematsu S, Akira S, Ogino Y, Kawauchi H, Unno N, Majima M. The inducible prostaglandin E synthase mPGES-1 regulates growth of endometrial tissues and angiogenesis in a mouse implantation model. Biomed Pharmacother 2011; 65(1): 77–84

[76]

Wolff EF, Mutlu L, Massasa EE, Elsworth JD, Eugene Redmond D Jr, Taylor HS. Endometrial stem cell transplantation in MPTP-exposed primates: an alternative cell source for treatment of Parkinson’s disease. J Cell Mol Med 2015; 19(1): 249–256

[77]

Santamaria X, Massasa EE, Feng Y, Wolff E, Taylor HS. Derivation of insulin producing cells from human endometrial stromal stem cells and use in the treatment of murine diabetes. Mol Ther 2011; 19(11): 2065–2071

[78]

Xie Q, Xiong X, Xiao N, He K, Chen M, Peng J, Su X, Mei H, Dai Y, Wei D, Lin G, Cheng L. Mesenchymal stem cells alleviate DHEA-induced polycystic ovary syndrome (PCOS) by inhibiting inflammation in mice. Stem Cells Int 2019; 2019: 9782373

[79]

Mohamed SA, Shalaby S, Brakta S, Elam L, Elsharoud A, Al-Hendy A. Umbilical cord blood mesenchymal stem cells as an infertility treatment for chemotherapy induced premature ovarian insufficiency. Biomedicines 2019; 7(1): 7

[80]

Song D, Zhong Y, Qian C, Zou Q, Ou J, Shi Y, Gao L, Wang G, Liu Z, Li H, Ding H, Wu H, Wang F, Wang J, Li H. Human umbilical cord mesenchymal stem cells therapy in cyclophosphamide-induced premature ovarian failure rat model. Biomed Res Int 2016; 2016: 2517514

[81]

Elfayomy AK, Almasry SM, El-Tarhouny SA, Eldomiaty MA. Human umbilical cord blood-mesenchymal stem cells transplantation renovates the ovarian surface epithelium in a rat model of premature ovarian failure: possible direct and indirect effects. Tissue Cell 2016; 48(4): 370–382

[82]

Zhang C. The roles of different stem cells in premature ovarian failure. Curr Stem Cell Res Ther 2020; 15(6): 473–481

[83]

Shi Q, Gao J, Jiang Y, Sun B, Lu W, Su M, Xu Y, Yang X, Zhang Y. Differentiation of human umbilical cord Wharton’s jelly-derived mesenchymal stem cells into endometrial cells. Stem Cell Res Ther 2017; 8(1): 246

[84]

Zhang L, Li Y, Guan CY, Tian S, Lv XD, Li JH, Ma X, Xia HF. Therapeutic effect of human umbilical cord-derived mesenchymal stem cells on injured rat endometrium during its chronic phase. Stem Cell Res Ther 2018; 9(1): 36

[85]

Xu L, Ding L, Wang L, Cao Y, Zhu H, Lu J, Li X, Song T, Hu Y, Dai J. Umbilical cord-derived mesenchymal stem cells on scaffolds facilitate collagen degradation via upregulation of MMP-9 in rat uterine scars. Stem Cell Res Ther 2017; 8(1): 84

[86]

Ding L, Yan G, Wang B, Xu L, Gu Y, Ru T, Cui X, Lei L, Liu J, Sheng X, Wang B, Zhang C, Yang Y, Jiang R, Zhou J, Kong N, Lu F, Zhou H, Zhao Y, Chen B, Hu Y, Dai J, Sun H. Transplantation of UC-MSCs on collagen scaffold activates follicles in dormant ovaries of POF patients with long history of infertility. Sci China Life Sci 2018; 61(12): 1554–1565

[87]

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(4): 1029–1036

[88]

Steigman SA, Ahmed A, Shanti RM, Tuan RS, Valim C, Fauza DO. Sternal repair with bone grafts engineered from amniotic mesenchymal stem cells. J Pediatr Surg 2009; 44(6): 1120–1126

[89]

Kunisaki SM, Fuchs JR, Kaviani A, Oh JT, LaVan DA, Vacanti JP, Wilson JM, Fauza DO. Diaphragmatic repair through fetal tissue engineering: a comparison between mesenchymal amniocyte- and myoblast-based constructs. J Pediatr Surg 2006; 41(1): 34–39

[90]

Xiao GY, Liu IH, Cheng CC, Chang CC, Lee YH, Cheng WT, Wu SC. Amniotic fluid stem cells prevent follicle atresia and rescue fertility of mice with premature ovarian failure induced by chemotherapy. PLoS One 2014; 9(9): e106538

[91]

Liu T, Huang Y, Guo L, Cheng W, Zou G. CD44+/CD105+ human amniotic fluid mesenchymal stem cells survive and proliferate in the ovary long-term in a mouse model of chemotherapy-induced premature ovarian failure. Int J Med Sci 2012; 9(7): 592–602

[92]

Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K, Bae YC, Jung JS. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem 2004; 14(4–6): 311–324

[93]

Damous LL, Nakamuta JS, Carvalho AE, Carvalho KC, Soares JM Jr, Simões MJ, Krieger JE, Baracat EC. Does adipose tissue-derived stem cell therapy improve graft quality in freshly grafted ovaries?. Reprod Biol Endocrinol 2015; 13(1): 108

[94]

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(4): 80

[95]

Kilic S, Yuksel B, Pinarli F, Albayrak A, Boztok B, Delibasi T. Effect of stem cell application on Asherman syndrome, an experimental rat model. J Assist Reprod Genet 2014; 31(8): 975–982

[96]

Ling L, Feng X, Wei T, Wang Y, Wang Y, Wang Z, Tang D, Luo Y, Xiong Z. Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation improves ovarian function in rats with premature ovarian insufficiency (POI) at least partly through a paracrine mechanism. Stem Cell Res Ther 2019; 10(1): 46

[97]

Feng X, Ling L, Zhang W, Liu X, Wang Y, Luo Y, Xiong Z. Effects of human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation in situ on primary ovarian insufficiency in SD rats. Reprod Sci 2020; 27(7): 1502–1512

[98]

Yin N, Wang Y, Lu X, Liu R, Zhang L, Zhao W, Yuan W, Luo Q, Wu H, Luan X, Zhang H. hPMSC transplantation restoring ovarian function in premature ovarian failure mice is associated with change of Th17/Tc17 and Th17/Treg cell ratios through the PI3K/Akt signal pathway. Stem Cell Res Ther 2018; 9(1): 37

[99]

Li H, Zhao W, Wang L, Luo Q, Yin N, Lu X, Hou Y, Cui J, Zhang H. Human placenta-derived mesenchymal stem cells inhibit apoptosis of granulosa cells induced by IRE1α pathway in autoimmune POF mice. Cell Biol Int 2019; 43(8): 899–909

[100]

Kim TH, Choi JH, Jun Y, Lim SM, Park S, Paek JY, Lee SH, Hwang JY, Kim GJ. 3D-cultured human placenta-derived mesenchymal stem cell spheroids enhance ovary function by inducing folliculogenesis. Sci Rep 2018; 8(1): 15313

[101]

Qu HM, Qu LP, Pan XZ, Mu LS. Upregulated miR-222 targets BCL2L11 and promotes apoptosis of mesenchymal stem cells in preeclampsia patients in response to severe hypoxia. Int J Clin Exp Pathol 2018; 11(1): 110–119

[102]

Manshadi MD, Navid S, Hoshino Y, Daneshi E, Noory P, Abbasi M. The effects of human menstrual blood stem cells-derived granulosa cells on ovarian follicle formation in a rat model of premature ovarian failure. Microsc Res Tech 2019; 82(6): 635–642

[103]

Domnina A, Novikova P, Obidina J, Fridlyanskaya I, Alekseenko L, Kozhukharova I, Lyublinskaya O, Zenin V, Nikolsky N. Human mesenchymal stem cells in spheroids improve fertility in model animals with damaged endometrium. Stem Cell Res Ther 2018; 9(1): 50

[104]

Zheng SX, Wang J, Wang XL, Ali A, Wu LM, Liu YS. Feasibility analysis of treating severe intrauterine adhesions by transplanting menstrual blood-derived stem cells. Int J Mol Med 2018; 41(4): 2201–2212

[105]

Zhang S, Li P, Yuan Z, Tan J. Platelet-rich plasma improves therapeutic effects of menstrual blood-derived stromal cells in rat model of intrauterine adhesion. Stem Cell Res Ther 2019; 10(1): 61

[106]

Yan Z, Guo F, Yuan Q, Shao Y, Zhang Y, Wang H, Hao S, Du X. Endometrial mesenchymal stem cells isolated from menstrual blood repaired epirubicin-induced damage to human ovarian granulosa cells by inhibiting the expression of Gadd45b in cell cycle pathway. Stem Cell Res Ther 2019; 10(1): 4

[107]

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(12): 2723–2729

[108]

Owen M, Friedenstein AJ. Stromal stem cells: marrow-derived osteogenic precursors. Ciba Found Symp 1988; 136: 42–60

[109]

Gao L, Huang Z, Lin H, Tian Y, Li P, Lin S. Bone marrow mesenchymal stem cells (BMSCs) restore functional endometrium in the rat model for severe Asherman syndrome. Reprod Sci 2019; 26(3): 436–444

[110]

Besikcioglu HE, Sarıbas GS, Ozogul C, Tiryaki M, Kilic S, Pınarlı FA, Gulbahar O. Determination of the effects of bone marrow derived mesenchymal stem cells and ovarian stromal stem cells on follicular maturation in cyclophosphamide induced ovarian failure in rats. Taiwan J Obstet Gynecol 2019; 58(1): 53–59

[111]

Guillot PV, Gotherstrom C, Chan J, Kurata H, Fisk NM. Human first-trimester fetal MSC express pluripotency markers and grow faster and have longer telomeres than adult MSC. Stem Cells 2007; 25(3): 646–654

[112]

Wang J, Ju B, Pan C, Gu Y, Zhang Y, Sun L, Zhang B, Zhang Y. Application of bone marrow-derived mesenchymal stem cells in the treatment of intrauterine adhesions in rats. Cell Physiol Biochem 2016; 39(4): 1553–1560

[113]

Santamaria X, Cabanillas S, Cervelló 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(5): 1087–1096

[114]

Cocuzza M, Alvarenga C, Pagani R. The epidemiology and etiology of azoospermia. Clinics (São Paulo) 2013; 68(Suppl 1): 15–26

[115]

Vij SC, Sabanegh E Jr, Agarwal A. Biological therapy for non-obstructive azoospermia. Expert Opin Biol Ther 2018; 18(1): 19–23

[116]

Kanatsu-Shinohara M, Shinohara T. Spermatogonial stem cell self-renewal and development. Annu Rev Cell Dev Biol 2013; 29(1): 163–187

[117]

Brinster RL, Zimmermann JW. Spermatogenesis following male germ-cell transplantation. Proc Natl Acad Sci USA 1994; 91(24): 11298–11302

[118]

Nayernia K, Nolte J, Michelmann HW, Lee JH, Rathsack K, Drusenheimer N, Dev A, Wulf G, Ehrmann IE, Elliott DJ, Okpanyi V, Zechner U, Haaf T, Meinhardt A, Engel W. In vitro-differentiated embryonic stem cells give rise to male gametes that can generate offspring mice. Dev Cell 2006; 11(1): 125–132

[119]

Hayashi K, Ohta H, Kurimoto K, Aramaki S, Saitou M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 2011; 146(4): 519–532

[120]

Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, Welt CK; Endocrine Society. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2013; 98(12): 4565–4592

[121]

Chugh R, Ashour D, Garcia N, Park H, Takala H, Ismail N, McAllister J, Al-Hendy A, El Andaloussi A. Towards cell therapy of polycystic ovary syndrome (PCOS): human mesenchymal stem cells secretome inhibits androgen production by pcos theca cells. Cytotherapy 2019; 21(5): S81

[122]

Yin M, Wang X, Yao G, M, Liang M, Sun Y, Sun F. Transactivation of micrornA-320 by microRNA-383 regulates granulosa cell functions by targeting E2F1 and SF-1 proteins. J Biol Chem 2014; 289(26): 18239–18257

[123]

Verkauf BS. Incidence, symptoms, and signs of endometriosis in fertile and infertile women. J Fla Med Assoc 1987; 74(9): 671–675

[124]

Practice Committee of the American Society for Reproductive Medicine. Endometriosis and infertility: a committee opinion. Fertil Steril 2012; 98(3): 591–598

[125]

Macer ML, Taylor HS. Endometriosis and infertility: a review of the pathogenesis and treatment of endometriosis-associated infertility. Obstet Gynecol Clin North Am 2012; 39(4): 535–549

[126]

Liu Y, Kodithuwakku SP, Ng PY, Chai J, Ng EH, Yeung WS, Ho PC, Lee KF. Excessive ovarian stimulation up-regulates the Wnt-signaling molecule DKK1 in human endometrium and may affect implantation: an in vitro co-culture study. Hum Reprod 2010; 25(2): 479–490

[127]

Sallam HN, Garcia-Velasco JA, Dias S, Arici A. Long-term pituitary down-regulation before in vitro fertilization (IVF) for women with endometriosis. Cochrane Database Syst Rev 2006; 2006(1): CD004635

[128]

Benschop L, Farquhar C, van der Poel N, Heineman MJ. Interventions for women with endometrioma prior to assisted reproductive technology. Cochrane Database Syst Rev 2010; (11): CD008571

[129]

European Society for Human Reproduction, Embryology (ESHRE) Guideline Group on POI, Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B, Cifkova R, de Muinck Keizer-Schrama S, Hogervorst E, Janse F, Liao L, Vlaisavljevic V, Zillikens C, Vermeulen N. ESHRE Guideline: management of women with premature ovarian insufficiency. Hum Reprod 2016; 31(5): 926–937

[130]

Kawamura K, Cheng Y, Suzuki N, Deguchi M, Sato Y, Takae S, Ho CH, Kawamura N, Tamura M, Hashimoto S, Sugishita Y, Morimoto Y, Hosoi Y, Yoshioka N, Ishizuka B, Hsueh AJ. Hippo signaling disruption and Akt stimulation of ovarian follicles for infertility treatment. Proc Natl Acad Sci USA 2013; 110(43): 17474–17479

[131]

Asherman JG. Traumatic intra-uterine adhesions. J Obstet Gynaecol Br Emp 1950; 57(6): 892–896

[132]

Schenker JG, Margalioth EJ. Intrauterine adhesions: an updated appraisal. Fertil Steril 1982; 37(5): 593–610

[133]

Valle RF, Sciarra JJ. Intrauterine adhesions: hysteroscopic diagnosis, classification, treatment, and reproductive outcome. Am J Obstet Gynecol 1988; 158(6): 1459–1470

[134]

Sugimoto O. Diagnostic and therapeutic hysteroscopy for traumatic intrauterine adhesions. Am J Obstet Gynecol 1978; 131(5): 539–547

[135]

Zikopoulos KA, Kolibianakis EM, Platteau P, de Munck L, Tournaye H, Devroey P, Camus M. Live delivery rates in subfertile women with Asherman’s syndrome after hysteroscopic adhesiolysis using the resectoscope or the Versapoint system. Reprod Biomed Online 2004; 8(6): 720–725

[136]

Alawadhi F, Du H, Cakmak H, Taylor HS. Bone marrow-derived stem cell (BMDSC) transplantation improves fertility in a murine model of Asherman’s syndrome. PLoS One 2014; 9(5): e96662

[137]

Gan L, Duan H, Xu Q, Tang YQ, Li JJ, Sun FQ, Wang S. Human amniotic mesenchymal stromal cell transplantation improves endometrial regeneration in rodent models of intrauterine adhesions. Cytotherapy 2017; 19(5): 603–616

[138]

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(2): 93–98

[139]

Vizoso FJ, Eiro N, Cid S, Schneider J, Perez-Fernandez R. Mesenchymal stem cell secretome: toward cell-free therapeutic strategies in regenerative medicine. Int J Mol Sci 2017; 18(9): 1852

[140]

Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant 2011; 20(1): 5–14

[141]

Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells—current trends and future prospective. Biosci Rep 2015; 35(2): e00191

[142]

Sneddon JB, Tang Q, Stock P, Bluestone JA, Roy S, Desai T, Hebrok M. Stem cell therapies for treating diabetes: progress and remaining challenges. Cell Stem Cell 2018; 22(6): 810–823

[143]

Du H, Taylor HS. Stem cells and female reproduction. Reprod Sci 2009; 16(2): 126–139

[144]

Liu F, Hu S, Yang H, Li Z, Huang K, Su T, Wang S, Cheng K. Hyaluronic acid hydrogel integrated with mesenchymal stem cell-secretome to treat endometrial injury in a rat model of Asherman’s syndrome. Adv Healthc Mater 2019; 8(14): e1900411

[145]

Mendt M, Rezvani K, Shpall E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplant 2019; 54(Suppl 2): 789–792

[146]

Ullah M, Qiao Y, Concepcion W, Thakor AS. Stem cell-derived extracellular vesicles: role in oncogenic processes, bioengineering potential, and technical challenges. Stem Cell Res Ther 2019; 10(1): 347

[147]

Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan , Brigstock DR, Brisson A, Broekman ML, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DR, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FA, Coyle B, Crescitelli R, Criado MF, D’Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, Del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TA, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, El Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, Fricke F, Fuhrmann G, Gabrielsson S, Gámez-Valero A, Gardiner C, Gärtner K, Gaudin R, Gho YS, Giebel B, Gilbert C, Gimona M, Giusti I, Goberdhan DC, Görgens A, Gorski SM, Greening DW, Gross JC, Gualerzi A, Gupta GN, Gustafson D, Handberg A, Haraszti RA, Harrison P, Hegyesi H, Hendrix A, Hill AF, Hochberg FH,Hoffmann KF,Holder B,Holthofer H,Hosseinkhani B,Hu G,Huang Y,Huber V, Hunt S,Ibrahim AG,Ikezu T,Inal JM,Isin M, Ivanova A,Jackson HK,Jacobsen S,Jay SM,Jayachandran M,Jenster G,Jiang L,Johnson SM, Jones JC,Jong A,Jovanovic-Talisman T, Jung S,Kalluri R, Kano SI, Kaur S,Kawamura Y,Keller ET,Khamari D,Khomyakova E,Khvorova A,Kierulf P,Kim KP, Kislinger T,Klingeborn M,Klinke DJ 2nd,Kornek M, Kosanović MM,KovácsÁF, Krämer-Albers EM, Krasemann S,Krause M,Kurochkin IV,Kusuma GD, Kuypers S,Laitinen S,Langevin SM,Languino LR,Lannigan J,Lässer C,Laurent LC,Lavieu G, Lázaro-Ibáñez E, Le Lay S, Lee MS, Lee YXF,Lemos DS,Lenassi M, Leszczynska A,Li IT,Liao K, Libregts SF,Ligeti E,Lim R,Lim SK,Linē A,Linnemannstöns K, Llorente A,Lombard CA,Lorenowicz MJ,LörinczÁM,Lötvall J, Lovett J, Lowry MC,Loyer X, Lu Q,Lukomska B,Lunavat TR,Maas SL,Malhi H, Marcilla A,Mariani J,Mariscal J,Martens-Uzunova ES,Martin-Jaular L,Martinez MC,Martins VR,Mathieu M, Mathivanan S,Maugeri M,McGinnis LK,McVey MJ,Meckes DG Jr,Meehan KL, Mertens I,Minciacchi VR,Möller A,Møller Jørgensen M, Morales-Kastresana A,Morhayim J,Mullier F,Muraca M,Musante L, Mussack V,Muth DC,Myburgh KH,Najrana T,Nawaz M, Nazarenko I,Nejsum P,Neri C,Neri T,Nieuwland R,Nimrichter L,Nolan JP,Nolte-’t Hoen EN,Noren Hooten N,O’Driscoll L,O’Grady T,O’Loghlen A,Ochiya T,Olivier M, Ortiz A,Ortiz LA,Osteikoetxea X,Østergaard O,Ostrowski M,Park J,Pegtel DM,Peinado H, Perut F,Pfaffl MW,Phinney DG,Pieters BC,Pink RC, Pisetsky DS,Pogge von Strandmann E,Polakovicova I,Poon IK,Powell BH,Prada I, Pulliam L,Quesenberry P,Radeghieri A,Raffai RL,Raimondo S,Rak J,Ramirez MI,Raposo G, Rayyan MS,Regev-Rudzki N,Ricklefs FL,Robbins PD,Roberts DD, Rodrigues SC,Rohde E,Rome S, Rouschop KM,Rughetti A,Russell AE,Saá P,Sahoo S, Salas-Huenuleo E,Sánchez C,Saugstad JA,Saul MJ,Schiffelers RM,Schneider R,Schøyen TH,Scott A,Shahaj E, Sharma S,Shatnyeva O,Shekari F,Shelke GV,Shetty AK, Shiba K,Siljander PR,Silva AM,Skowronek A,Snyder OL 2nd, Soares RP, Sódar BW,Soekmadji C,Sotillo J,Stahl PD,Stoorvogel W,Stott SL,Strasser EF,Swift S, Tahara H,Tewari M,Timms K,Tiwari S,Tixeira R, Tkach M,Toh WS,Tomasini R,Torrecilhas AC,Tosar JP, Toxavidis V,Urbanelli L,Vader P,van Balkom BW,van der Grein SG,Van Deun J,van Herwijnen MJ,Van Keuren-Jensen K, van Niel G, van Royen ME,van Wijnen AJ,Vasconcelos MH,Vechetti IJ Jr,Veit TD, Vella LJ,Velot É,Verweij FJ,Vestad B,Viñas JL,Visnovitz T,Vukman KV,Wahlgren J,Watson DC,Wauben MH,Weaver A, Webber JP,Weber V,Wehman AM,Weiss DJ,Welsh JA, Wendt S,Wheelock AM, Wiener Z, Witte L, Wolfram J, Xagorari A, Xander P, Xu J, Yan X, Yáñez-Mó M, Yin H, Yuana Y, Zappulli V, Zarubova J, Žėkas V, Zhang JY, Zhao Z, Zheng L, Zheutlin AR, Zickler AM, Zimmermann P, Zivkovic AM, Zocco D, Zuba-Surma EK. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018; 7(1): 1535750

[148]

Zhao AG, Shah K, Cromer B, Sumer H. Mesenchymal stem cell-derived extracellular vesicles and their therapeutic potential. Stem Cells Int 2020; 2020: 8825771

[149]

Yu Y, Lin X, Wang Q, He M, Chau Y. Long-term therapeutic effect in nonhuman primate eye from a single injection of anti-VEGF controlled release hydrogel. Bioeng Transl Med 2019; 4(2): e10128

[150]

Kourembanas S. Exosomes: vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu Rev Physiol 2015; 77(1): 13–27

[151]

Qiu G, Zheng G, Ge M, Wang J, Huang R, Shu Q, Xu J. Functional proteins of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther 2019; 10(1): 359

[152]

Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013; 200(4): 373–383

[153]

Joo HS, Suh JH, Lee HJ, Bang ES, Lee JM. Current knowledge and future perspectives on mesenchymal stem cell-derived exosomes as a new therapeutic agent. Int J Mol Sci 2020; 21(3): 727

[154]

Abbaszadeh H, Ghorbani F, Derakhshani M, Movassaghpour A, Yousefi M. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles: a novel therapeutic paradigm. J Cell Physiol 2020; 235(2): 706–717

[155]

Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, Stolz DB, Watkins SC, Di YP, Leikauf GD, Kolls J, Riches DW, Deiuliis G, Kaminski N, Boregowda SV, McKenna DH, Ortiz LA. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun 2015; 6(1): 8472

[156]

Domniz N, Meirow D. Premature ovarian insufficiency and autoimmune diseases. Best Pract Res Clin Obstet Gynaecol 2019; 60: 42–55

[157]

De Vos M, Devroey P, Fauser BC. Primary ovarian insufficiency. Lancet 2010; 376(9744): 911–921

[158]

Huang B, Qian C, Ding C, Meng Q, Zou Q, Li H. Fetal liver mesenchymal stem cells restore ovarian function in premature ovarian insufficiency by targeting MT1. Stem Cell Res Ther 2019; 10(1): 362

[159]

Kim EH, Jeon BH, Kim J, Kim YM, Han Y, Ahn K, Cheong HK. Exposure to phthalates and bisphenol A are associated with atopic dermatitis symptoms in children: a time-series analysis. Environ Health 2017; 16(1): 24

[160]

Ford EA, Beckett EL, Roman SD, McLaughlin EA, Sutherland JM. Advances in human primordial follicle activation and premature ovarian insufficiency. Reproduction 2020; 159(1): R15–R29

[161]

Huhtaniemi I, Hovatta O, La Marca A, Livera G, Monniaux D, Persani L, Heddar A, Jarzabek K, Laisk-Podar T, Salumets A, Tapanainen JS, Veitia RA, Visser JA, Wieacker P, Wolczynski S, Misrahi M. Advances in the molecular pathophysiology, genetics, and treatment of primary ovarian insufficiency. Trends Endocrinol Metab 2018; 29(6): 400–419

[162]

Wesevich V, Kellen AN, Pal L. Recent advances in understanding primary ovarian insufficiency. F1000 Res 2020; 9: F1000 Faculty Rev-1101

[163]

Caburet S, Arboleda VA, Llano E, Overbeek PA, Barbero JL, Oka K, Harrison W, Vaiman D, Ben-Neriah Z, García-Tuñón I, Fellous M, Pendás AM, Veitia RA, Vilain E. Mutant cohesin in premature ovarian failure. N Engl J Med 2014; 370(10): 943–949

[164]

Bouilly J, Beau I, Barraud S, Bernard V, Azibi K, Fagart J, Fèvre A, Todeschini AL, Veitia RA, Beldjord C, Delemer B, Dodé C, Young J, Binart N. Identification of multiple gene mutations accounts for a new genetic architecture of primary ovarian insufficiency. J Clin Endocrinol Metab 2016; 101(12): 4541–4550

[165]

Jaillard S, Bell K, Akloul L, Walton K, McElreavy K, Stocker WA, Beaumont M, Harrisson C, Jääskeläinen T, Palvimo JJ, Robevska G, Launay E, Satié AP, Listyasari N, Bendavid C, Sreenivasan R, Duros S, van den Bergen J, Henry C, Domin-Bernhard M, Cornevin L, Dejucq-Rainsford N, Belaud-Rotureau MA, Odent S, Ayers KL, Ravel C, Tucker EJ, Sinclair AH. New insights into the genetic basis of premature ovarian insufficiency: novel causative variants and candidate genes revealed by genomic sequencing. Maturitas 2020; 141: 9–19

[166]

Liu T, Liu Y, Huang Y, Chen J, Yu Z, Chen C, Lai L. miR-15b induces premature ovarian failure in mice via inhibition of α-Klotho expression in ovarian granulosa cells. Free Radic Biol Med 2019; 141: 383–392

[167]

Persani L, Rossetti R, Cacciatore C, Fabre S. Genetic defects of ovarian TGF-β-like factors and premature ovarian failure. J Endocrinol Invest 2011; 34(3): 244–251

[168]

Grosbois J, Demeestere I. Dynamics of PI3K and Hippo signaling pathways during in vitro human follicle activation. Hum Reprod 2018; 33(9): 1705–1714

[169]

Dragojević-Dikić S, Marisavljević D, Mitrović A, Dikić S, Jovanović T, Janković-Raznatović S. An immunological insight into premature ovarian failure (POF). Autoimmun Rev 2010; 9(11): 771–774

[170]

Coulam CB, Stern JJ. Immunology of ovarian failure. Am J Reprod Immunol 1991; 25(4): 169–174

[171]

Shen M, Jiang Y, Guan Z, Cao Y, Li L, Liu H, Sun SC. Protective mechanism of FSH against oxidative damage in mouse ovarian granulosa cells by repressing autophagy. Autophagy 2017; 13(8): 1364–1385

[172]

Huang B, Lu J, Ding C, Zou Q, Wang W, Li H. Exosomes derived from human adipose mesenchymal stem cells improve ovary function of premature ovarian insufficiency by targeting SMAD. Stem Cell Res Ther 2018; 9(1): 216

[173]

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(4): 527–533

[174]

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(1): 250

[175]

Zhang S, Huang B, Su P, Chang Q, Li P, Song A, Zhao X, Yuan Z, Tan J. Concentrated exosomes from menstrual blood-derived stromal cells improves ovarian activity in a rat model of premature ovarian insufficiency. Stem Cell Res Ther 2021; 12(1): 178

[176]

Xiao GY, Cheng CC, Chiang YS, Cheng WT, Liu IH, Wu SC. Exosomal miR-10a derived from amniotic fluid stem cells preserves ovarian follicles after chemotherapy. Sci Rep 2016; 6(1): 23120

[177]

Sun L, Li D, Song K, Wei J, Yao S, Li Z, Su X, Ju X, Chao L, Deng X, Kong B, Li L. Exosomes derived from human umbilical cord mesenchymal stem cells protect against cisplatin-induced ovarian granulosa cell stress and apoptosis in vitro. Sci Rep 2017; 7(1): 2552

[178]

Boecker W, van Horn L, Stenman G, Stürken C, Schumacher U, Loening T, Liesenfeld L, Korsching E, Gläser D, Tiemann K, Buchwalow I. Spatially correlated phenotyping reveals K5-positive luminal progenitor cells and p63-K5/14-positive stem cell-like cells in human breast epithelium. Lab Invest 2018; 98(8): 1065–1075

[179]

Thabet E, Yusuf A, Abdelmonsif DA, Nabil I, Mourad G, Mehanna RA. Extracellular vesicles miRNA-21: a potential therapeutic tool in premature ovarian dysfunction. Mol Hum Reprod 2020; 26(12): 906–919

[180]

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(1): 360

[181]

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

[182]

Liu C, Yin H, Jiang H, Du X, Wang C, Liu Y, Li Y, Yang Z. Extracellular vesicles derived from mesenchymal stem cells recover fertility of premature ovarian insufficiency mice and the effects on their offspring. Cell Transplant 2020; 29: 963689720923575

[183]

Norman RJ, Wu R, Stankiewicz MT. 4: Polycystic ovary syndrome. Med J Aust 2004; 180(3): 132–137

[184]

Blázquez R, Sánchez-Margallo FM, Álvarez V, Matilla E, Hernández N, Marinaro F, Gómez-Serrano M, Jorge I, Casado JG, Macías-García B. Murine embryos exposed to human endometrial MSCs-derived extracellular vesicles exhibit higher VEGF/PDGF AA release, increased blastomere count and hatching rates. PLoS One 2018; 13(4): e0196080

[185]

Marinaro F, Macías-García B, Sánchez-Margallo FM, Blázquez R, Álvarez V, Matilla E, Hernández N, Gómez-Serrano M, Jorge I, Vázquez J, González-Fernández L, Pericuesta E, Gutiérrez-Adán A, Casado JG. Extracellular vesicles derived from endometrial human mesenchymal stem cells enhance embryo yield and quality in an aged murine model. Biol Reprod 2019; 100(5): 1180–1192

[186]

The Lancet Diabetes Endocrinology. Empowering women with PCOS. Lancet Diabetes Endocrinol 2019; 7(10): 737

[187]

Escobar-Morreale HF. Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nat Rev Endocrinol 2018; 14(5): 270–284

[188]

Liang X, Zhang L, Wang S, Han Q, Zhao RC. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J Cell Sci 2016; 129(11): 2182–2189

[189]

Balen AH, Morley LC, Misso M, Franks S, Legro RS, Wijeyaratne CN, Stener-Victorin E, Fauser BC, Norman RJ, Teede H. The management of anovulatory infertility in women with polycystic ovary syndrome: an analysis of the evidence to support the development of global WHO guidance. Hum Reprod Update 2016; 22(6): 687–708

[190]

Madkour A, Bouamoud N, Kaarouch I, Louanjli N, Saadani B, Assou S, Aboulmaouahib S, Sefrioui O, Amzazi S, Copin H, Benkhalifa M. Follicular fluid and supernatant from cultured cumulus-granulosa cells improve in vitro maturation in patients with polycystic ovarian syndrome. Fertil Steril 2018; 110(4): 710–719

[191]

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(1): 3804–3813

[192]

Salazar CA, Isaacson K, Morris S. A comprehensive review of Asherman’s syndrome: causes, symptoms and treatment options. Curr Opin Obstet Gynecol 2017; 29(4): 249–256

[193]

March CM. Asherman’s syndrome. Semin Reprod Med 2011; 29(2): 83–94

[194]

Lo ST, Ramsay P, Pierson R, Manconi F, Munro MG, Fraser IS. Endometrial thickness measured by ultrasound scan in women with uterine outlet obstruction due to intrauterine or upper cervical adhesions. Hum Reprod 2008; 23(2): 306–309

[195]

Xiao S, Wan Y, Xue M, Zeng X, Xiao F, Xu D, Yang X, Zhang P, Sheng W, Xu J, Zhou S. Etiology, treatment, and reproductive prognosis of women with moderate-to-severe intrauterine adhesions. Int J Gynaecol Obstet 2014; 125(2): 121–124

[196]

Bai X, Liu J, Cao S, Wang L. Mechanisms of endometrial fibrosis and the potential application of stem cell therapy. Discov Med 2019; 27(150): 267–279

[197]

Santamaria X, Isaacson K, Simón C. Asherman’s syndrome: it may not be all our fault. Hum Reprod 2018; 33(8): 1374–1380

[198]

Salma U, Xue M, Ali Sheikh MS, Guan X, Xu B, Zhang A, Huang L, Xu D. Role of transforming growth factor-β1 and Smads signaling pathway in intrauterine adhesion. Mediators Inflamm 2016; 2016: 4158287

[199]

Wang X, Ma N, Sun Q, Huang C, Liu Y, Luo X. Elevated NF-κB signaling in Asherman syndrome patients and animal models. Oncotarget 2017; 8(9): 15399–15406

[200]

Xue X, Chen Q, Zhao G, Zhao JY, Duan Z, Zheng PS. The overexpression of TGF-β and CCN2 in intrauterine adhesions involves the NF-κB signaling pathway. PLoS One 2015; 10(12): e0146159

[201]

Zhu HY, Ge TX, Pan YB, Zhang SY. Advanced role of hippo signaling in endometrial fibrosis: implications for intrauterine adhesion. Chin Med J (Engl) 2017; 130(22): 2732–2737

[202]

Akhmetshina A, Palumbo K, Dees C, Bergmann C, Venalis P, Zerr P, Horn A, Kireva T, Beyer C, Zwerina J, Schneider H, Sadowski A, Riener MO, MacDougald OA, Distler O, Schett G, Distler JH. Activation of canonical Wnt signalling is required for TGF-β-mediated fibrosis. Nat Commun 2012; 3(1): 735

[203]

Liu L, Chen G, Chen T, Shi W, Hu H, Song K, Huang R, Cai H, He Y. si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway. Stem Cell Res Ther 2020; 11(1): 1–17

[204]

Chen G, Liu L, Sun J, Zeng L, Cai H, He Y. Foxf2 and Smad6 co-regulation of collagen 5A2 transcription is involved in the pathogenesis of intrauterine adhesion. J Cell Mol Med 2020; 24(5): 2802–2818

[205]

Leung RKK, Lin Y, Liu Y. Recent advances in understandings towards pathogenesis and treatment for intrauterine adhesion and disruptive insights from single-cell analysis. Reprod Sci 2021; 28(7): 1812–1826

[206]

Liu M, Zhao D, Wu X, Guo S, Yan L, Zhao S, Li H, Wang Y, Rong F. miR-466 and NUS1 regulate the AKT/nuclear factor kappa B (NFκB) signaling pathway in intrauterine adhesions in a rat model. Med Sci Monit 2019; 25: 4094–4103

[207]

Xu Q, Duan H, Gan L, Liu X, Chen F, Shen X, Tang YQ, Wang S. MicroRNA-1291 promotes endometrial fibrosis by regulating the ArhGAP29-RhoA/ROCK1 signaling pathway in a murine model. Mol Med Rep 2017; 16(4): 4501–4510

[208]

Zhu H, Pan Y, Jiang Y, Li J, Zhang Y, Zhang S. Activation of the Hippo/TAZ pathway is required for menstrual stem cells to suppress myofibroblast and inhibit transforming growth factor β signaling in human endometrial stromal cells. Hum Reprod 2019; 34(4): 635–645

[209]

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(6): 1266–1275

[210]

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

[211]

Perrini C, Strillacci MG, Bagnato A, Esposti P, Marini MG, Corradetti B, Bizzaro D, Idda A, Ledda S, Capra E, Pizzi F, Lange-Consiglio A, Cremonesi F. 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(1): 169

[212]

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(1): 225

[213]

Saribas GS, Ozogul C, Tiryaki M, Alpaslan Pinarli F, Hamdemir Kilic S. Effects of uterus derived mesenchymal stem cells and their exosomes on Asherman’s syndrome. Acta Histochem 2020; 122(1): 151465

[214]

Chang Y, Liu Y, Li X. Exosomes derived from human umbilical cord mesenchymal stem cells promote proliferation of endometrial stromal cell. Fertil Steril 2020; 114(3): e530

[215]

Aworunse OS, Adeniji O, Oyesola OL, Isewon I, Oyelade J, Obembe OO. Genomic interventions in medicine. Bioinform Biol Insights 2018; 12: 1177932218816100

[216]

Goetz LH, Schork NJ. Personalized medicine: motivation, challenges, and progress. Fertil Steril 2018; 109(6): 952–963

[217]

Zhang PY, Yu Y. Precise personalized medicine in gynecology cancer and infertility. Front Cell Dev Biol 2020; 7: 382

[218]

Ferlin A, Arredi B, Foresta C. Genetic causes of male infertility. Reprod Toxicol 2006; 22(2): 133–141

[219]

Bracke A, Peeters K, Punjabi U, Hoogewijs D, Dewilde S. A search for molecular mechanisms underlying male idiopathic infertility. Reprod Biomed Online 2018; 36(3): 327–339

[220]

Ventimiglia E, Montorsi F, Salonia A. Comorbidities and male infertility: a worrisome picture. Curr Opin Urol 2016; 26(2): 146–151

[221]

Jungwirth A, Giwercman A, Tournaye H, Diemer T, Kopa Z, Dohle G, Krausz C; European Association of Urology Working Group on Male Infertility. European Association of Urology guidelines on Male Infertility: the 2012 update. Eur Urol 2012; 62(2): 324–332

[222]

Oud MS, Volozonoka L, Smits RM, Vissers LELM, Ramos L, Veltman JA. A systematic review and standardized clinical validity assessment of male infertility genes. Hum Reprod 2019; 34(5): 932–941

[223]

Krausz C, Escamilla AR, Chianese C. Genetics of male infertility: from research to clinic. Reproduction 2015; 150(5): R159–R174

[224]

Cariati F, D’Argenio V, Tomaiuolo R. The evolving role of genetic tests in reproductive medicine. J Transl Med 2019; 17(1): 267

[225]

Krausz C, Degl’Innocenti S. Y chromosome and male infertility: update, 2006. Front Biosci 2006; 11(1): 3049–3061

[226]

Kim IW, Khadilkar AC, Ko EY, Sabanegh ES Jr. 47, XYY syndrome and male infertility. Rev Urol 2013; 15(4): 188–196

[227]

Krausz C, Hoefsloot L, Simoni M, Tüttelmann F; European Academy of Andrology; European Molecular Genetics Quality Network. EAA/EMQN best practice guidelines for molecular diagnosis of Y-chromosomal microdeletions: state-of-the-art 2013. Andrology 2014; 2(1): 5–19

[228]

Stuppia L, Gatta V, Calabrese G, Guanciali Franchi P, Morizio E, Bombieri C, Mingarelli R, Sforza V, Frajese G, Tenaglia R, Palka G. A quarter of men with idiopathic oligo-azoospermia display chromosomal abnormalities and microdeletions of different types in interval 6 of Yq11. Hum Genet 1998; 102(5): 566–570

[229]

Patsalis PC, Sismani C, Quintana-Murci L, Taleb-Bekkouche F, Krausz C, McElreavey K. Effects of transmission of Y chromosome AZFc deletions. Lancet 2002; 360(9341): 1222–1224

[230]

Asero P, Calogero AE, Condorelli RA, Mongioi’ L, Vicari E, Lanzafame F, Crisci R, La Vignera S. Relevance of genetic investigation in male infertility. J Endocrinol Invest 2014; 37(5): 415–427

[231]

Kuroda-Kawaguchi T, Skaletsky H, Brown LG, Minx PJ, Cordum HS, Waterston RH, Wilson RK, Silber S, Oates R, Rozen S, Page DC. The AZFc region of the Y chromosome features massive palindromes and uniform recurrent deletions in infertile men. Nat Genet 2001; 29(3): 279–286

[232]

Ferlin A, Garolla A, Foresta C. Chromosome abnormalities in sperm of individuals with constitutional sex chromosomal abnormalities. Cytogenet Genome Res 2005; 111(3–4): 310–316

[233]

Schultz N, Hamra FK, Garbers DL. A multitude of genes expressed solely in meiotic or postmeiotic spermatogenic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci USA 2003; 100(21): 12201–12206

[234]

Mastantuoni E, Saccone G, Al-Kouatly HB, Paternoster M, D’Alessandro P, Arduino B, Carbone L, Esposito G, Raffone A, De Vivo V, Maruotti GM, Berghella V, Zullo F. Expanded carrier screening: a current perspective. Eur J Obstet Gynecol Reprod Biol 2018; 230: 41–54

[235]

Morin SJ, Eccles J, Iturriaga A, Zimmerman RS. Translocations, inversions and other chromosome rearrangements. Fertil Steril 2017; 107(1): 19–26

[236]

Folsom LJ, Fuqua JS. Reproductive issues in women with turner syndrome. Endocrinol Metab Clin North Am 2015; 44(4): 723–737

[237]

Oktay K, Bedoschi G, Berkowitz K, Bronson R, Kashani B, McGovern P, Pal L, Quinn G, Rubin K. Fertility preservation in women with Turner syndrome: a comprehensive review and practical guidelines. J Pediatr Adolesc Gynecol 2016; 29(5): 409–416

[238]

Chen M, Wei S, Hu J, Quan S. Can comprehensive chromosome screening technology improve IVF/ICSI outcomes? A meta-analysis.. PLoS One 2015; 10(10): e0140779

[239]

Man L, Lekovich J, Rosenwaks Z, Gerhardt J. Fragile X-associated diminished ovarian reserve and primary ovarian insufficiency from molecular mechanisms to clinical manifestations. Front Mol Neurosci 2017; 10: 290

[240]

Hoyos LR, Thakur M. Fragile X premutation in women: recognizing the health challenges beyond primary ovarian insufficiency. J Assist Reprod Genet 2017; 34(3): 315–323

[241]

Rossetti R, Ferrari I, Bonomi M, Persani L. Genetics of primary ovarian insufficiency. Clin Genet 2017; 91(2): 183–198

[242]

Stavljenić-Rukavina A. 1. Prenatal diagnosis of chromosomal disorders—molecular aspects. EJIFCC 2008; 19(1): 2–6

[243]

Bennett RL. The family medical history as a tool in preconception consultation. J Community Genet 2012; 3(3): 175–183

[244]

Mathijssen IB, Holtkamp KCA, Ottenheim CPE, van Eeten-Nijman JMC, Lakeman P, Meijers-Heijboer H, van Maarle MC, Henneman L. Preconception carrier screening for multiple disorders: evaluation of a screening offer in a Dutch founder population. Eur J Hum Genet 2018; 26(2): 166–175

[245]

Dorney E, Black KI. Preconception care. Aust J Gen Pract 2018; 47(7): 424–429

[246]

Elce A, Boccia A, Cardillo G, Giordano S, Tomaiuolo R, Paolella G, Castaldo G. Three novel CFTR polymorphic repeats improve segregation analysis for cystic fibrosis. Clin Chem 2009; 55(7): 1372–1379

[247]

Cariati F, Savarese M, D’Argenio V, Salvatore F, Tomaiuolo R. The SEeMORE strategy: single-tube electrophoresis analysis-based genotyping to detect monogenic diseases rapidly and effectively from conception until birth. Clin Chem Lab Med 2017; 56(1): 40–50

[248]

Allyse M, Minear MA, Berson E, Sridhar S, Rote M, Hung A, Chandrasekharan S. Non-invasive prenatal testing: a review of international implementation and challenges. Int J Womens Health 2015; 7: 113–126

[249]

Chiu EKL, Hui WWI, Chiu RWK. cfDNA screening and diagnosis of monogenic disorders—where are we heading?. Prenat Diagn 2018; 38(1): 52–58

[250]

Saba L, Masala M, Capponi V, Marceddu G, Massidda M, Rosatelli MC. Non-invasive prenatal diagnosis of beta-thalassemia by semiconductor sequencing: a feasibility study in the sardinian population. Eur J Hum Genet 2017; 25(5): 600–607

[251]

New MI, Tong YK, Yuen T, Jiang P, Pina C, Chan KC, Khattab A, Liao GJ, Yau M, Kim SM, Chiu RW, Sun L, Zaidi M, Lo YM. Noninvasive prenatal diagnosis of congenital adrenal hyperplasia using cell-free fetal DNA in maternal plasma. J Clin Endocrinol Metab 2014; 99(6): E1022–E1030

[252]

Haahr MK, Jensen CH, Toyserkani NM, Andersen DC, Damkier P, Sørensen JA, Lund L, Sheikh SP. Safety and potential effect of a single intracavernous injection of autologous adipose-derived regenerative cells in patients with erectile dysfunction following radical prostatectomy: an open-label phase I clinical trial. EBioMedicine 2016; 5: 204–210

[253]

Mashayekhi M, Mirzadeh E, Chekini Z, Ahmadi F, Eftekhari-Yazdi P, Vesali S, Madani T, Aghdami N. Evaluation of safety, feasibility and efficacy of intra-ovarian transplantation of autologous adipose derived mesenchymal stromal cells in idiopathic premature ovarian failure patients: non-randomized clinical trial, phase I, first in human. J Ovarian Res 2021; 14(1): 5

[254]

Igboeli P, El Andaloussi A, Sheikh U, Takala H, ElSharoud A, McHugh A, Gavrilova-Jordan L, Levy S, Al-Hendy A. Intraovarian injection of autologous human mesenchymal stem cells increases estrogen production and reduces menopausal symptoms in women with premature ovarian failure: two case reports and a review of the literature. J Med Case Reports 2020; 14(1): 108

[255]

Cheng YC, Takagi M, Milbourne A, Champlin RE, Ueno NT. Phase II study of gonadotropin-releasing hormone analog for ovarian function preservation in hematopoietic stem cell transplantation patients. Oncologist 2012; 17(2): 233–238

[256]

Cao Y, Sun H, Zhu H, Zhu X, Tang X, Yan G, Wang J, Bai D, Wang J, Wang L, Zhou Q, Wang H, Dai C, Ding L, Xu B, Zhou Y, Hao J, Dai J, Hu Y. Allogeneic cell therapy using umbilical cord MSCs on collagen scaffolds for patients with recurrent uterine adhesion: a phase I clinical trial. Stem Cell Res Ther 2018; 9(1): 192

[257]

Yang W, Zhang J, Xu B, He Y, Liu W, Li J, Zhang S, Lin X, Su D, Wu T, Li J. HucMSC-derived exosomes mitigate the age-related retardation of fertility in female mice. Mol Ther 2020; 28(4): 1200–1213

[258]

Spinosa M, Lu G, Su G, Bontha SV, Gehrau R, Salmon MD, Smith JR, Weiss ML, Mas VR, Upchurch GR Jr, Sharma AK. Human mesenchymal stromal cell-derived extracellular vesicles attenuate aortic aneurysm formation and macrophage activation via microRNA-147. FASEB J 2018; 32(11): 6038–6050

[259]

Lo Sicco C, Reverberi D, Balbi C, Ulivi V, Principi E, Pascucci L, Becherini P, Bosco MC, Varesio L, Franzin C, Pozzobon M, Cancedda R, Tasso R. Mesenchymal stem cell-derived extracellular vesicles as mediators of anti-inflammatory effects: endorsement of macrophage polarization. Stem Cells Transl Med 2017; 6(3): 1018–1028

[260]

Ding C, Zhu L, Shen H, Lu J, Zou Q, Huang C, Li H, Huang B. Exosomal miRNA-17-5p derived from human umbilical cord mesenchymal stem cells improves ovarian function in premature ovarian insufficiency by regulating SIRT7. Stem Cells 2020; 38(9): 1137–1148

[261]

Bodart-Santos V, de Carvalho LRP, de Godoy MA, Batista AF, Saraiva LM, Lima LG, Abreu CA, De Felice FG, Galina A, Mendez-Otero R, Ferreira ST. Extracellular vesicles derived from human Wharton’s jelly mesenchymal stem cells protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. Stem Cell Res Ther 2019; 10(1): 332

[262]

Chaubey S, Thueson S, Ponnalagu D, Alam MA, Gheorghe CP, Aghai Z, Singh H, Bhandari V. Early gestational mesenchymal stem cell secretome attenuates experimental bronchopulmonary dysplasia in part via exosome-associated factor TSG-6. Stem Cell Res Ther 2018; 9(1): 173

[263]

Yang C, Lim W, Park J, Park S, You S, Song G. Anti-inflammatory effects of mesenchymal stem cell-derived exosomal microRNA-146a-5p and microRNA-548e-5p on human trophoblast cells. Mol Hum Reprod 2019; 25(11): 755–771

[264]

Ti D, Hao H, Tong C, Liu J, Dong L, Zheng J, Zhao Y, Liu H, Fu X, Han W. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J Transl Med 2015; 13(1): 308

[265]

Alzubi MA, Sohal SS, Sriram M, Turner TH, Zot P, Idowu M, Harrell JC. Quantitative assessment of breast cancer liver metastasis expansion with patient-derived xenografts. Clin Exp Metastasis 2019; 36(3): 257–269

[266]

Anderson JD, Johansson HJ, Graham CS, Vesterlund M, Pham MT, Bramlett CS, Montgomery EN, Mellema MS, Bardini RL, Contreras Z, Hoon M, Bauer G, Fink KD, Fury B, Hendrix KJ, Chedin F, El-Andaloussi S, Hwang B, Mulligan MS, Lehtiö J, Nolta JA. Comprehensive proteomic analysis of mesenchymal stem cell exosomes reveals modulation of angiogenesis via nuclear factor-kappaB signaling. Stem Cells 2016; 34(3): 601–613

[267]

Lopatina T, Bruno S, Tetta C, Kalinina N, Porta M, Camussi G. Platelet-derived growth factor regulates the secretion of extracellular vesicles by adipose mesenchymal stem cells and enhances their angiogenic potential. Cell Commun Signal 2014; 12(1): 26

[268]

Park KS, Svennerholm K, Shelke GV, Bandeira E, Lässer C, Jang SC, Chandode R, Gribonika I, Lötvall J. Mesenchymal stromal cell-derived nanovesicles ameliorate bacterial outer membrane vesicle-induced sepsis via IL-10. Stem Cell Res Ther 2019; 10(1): 231

[269]

Gonzalez-King H, García NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepúlveda P. Hypoxia inducible factor-1α potentiates jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem Cells 2017; 35(7): 1747–1759

[270]

Gong M, Yu B, Wang J, Wang Y, Liu M, Paul C, Millard RW, Xiao DS, Ashraf M, Xu M. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget 2017; 8(28): 45200–45212

[271]

Zhang B, Wang M, Gong A, Zhang X, Wu X, Zhu Y, Shi H, Wu L, Zhu W, Qian H, Xu W. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells 2015; 33(7): 2158–2168

[272]

Baek G, Choi H, Kim Y, Lee HC, Choi C. Mesenchymal stem cell-derived extracellular vesicles as therapeutics and as a drug delivery platform. Stem Cells Transl Med 2019; 8(9): 880–886

[273]

Liu L, Liu Y, Feng C, Chang J, Fu R, Wu T, Yu F, Wang X, Xia L, Wu C, Fang B. Lithium-containing biomaterials stimulate bone marrow stromal cell-derived exosomal miR-130a secretion to promote angiogenesis. Biomaterials 2019; 192: 523–536

[274]

Yang M, Lin L, Sha C, Li T, Zhao D, Wei H, Chen Q, Liu Y, Chen X, Xu W, Li Y, Zhu X. Bone marrow mesenchymal stem cell-derived exosomal miR-144-5p improves rat ovarian function after chemotherapy-induced ovarian failure by targeting PTEN. Lab Invest 2020; 100(3): 342–352

[275]

Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, Gao L, Xie J, Xu B. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc Res 2019; 115(7): 1205–1216

[276]

Eirin A, Zhu XY, Puranik AS, Woollard JR, Tang H, Dasari S, Lerman A, van Wijnen AJ, Lerman LO. Comparative proteomic analysis of extracellular vesicles isolated from porcine adipose tissue-derived mesenchymal stem/stromal cells. Sci Rep 2016; 6(1): 36120

[277]

Vrijsen KR, Maring JA, Chamuleau SA, Verhage V, Mol EA, Deddens JC, Metz CH, Lodder K, van Eeuwijk EC, van Dommelen SM, Doevendans PA, Smits AM, Goumans MJ, Sluijter JP. Exosomes from cardiomyocyte progenitor cells and mesenchymal stem cells stimulate angiogenesis via EMMPRIN. Adv Healthc Mater 2016; 5(19): 2555–2565

[278]

Crain SK, Robinson SR, Thane KE, Davis AM, Meola DM, Barton BA, Yang VK, Hoffman AM. Extracellular vesicles from Wharton’s jelly mesenchymal stem cells suppress CD4 expressing T cells through transforming growth factor beta and adenosine signaling in a canine model. Stem Cells Dev 2019; 28(3): 212–226

[279]

Mardpour S, Hamidieh AA, Taleahmad S, Sharifzad F, Taghikhani A, Baharvand H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J Cell Physiol 2019; 234(6): 8249–8258

[280]

Merino-González C, Zuñiga FA, Escudero C, Ormazabal V, Reyes C, Nova-Lamperti E, Salomón C, Aguayo C. Mesenchymal stem cell-derived extracellular vesicles promote angiogenesis: potencial clinical application. Front Physiol 2016; 7: 24

[281]

Tsuji K, Kitamura S, Wada J. Immunomodulatory and regenerative effects of mesenchymal stem cell-derived extracellular vesicles in renal diseases. Int J Mol Sci 2020; 21(3): 756

[282]

Paduano F, Marrelli M, Palmieri F, Tatullo M. CD146 expression influences periapical cyst mesenchymal stem cell properties. Stem Cell Rev Rep 2016; 12(5): 592–603

[283]

Momose T, Miyaji H, Kato A, Ogawa K, Yoshida T, Nishida E, Murakami S, Kosen Y, Sugaya T, Kawanami M. Collagen hydrogel scaffold and fibroblast growth factor-2 accelerate periodontal healing of class II furcation defects in dog. Open Dent J 2016; 10(1): 347–359

[284]

Cervelló I, Gil-Sanchis C, Santamaría X, Cabanillas S, Díaz A, Faus A, Pellicer A, Simón C. Human CD133(+) bone marrow-derived stem cells promote endometrial proliferation in a murine model of Asherman syndrome. Fertil Steril 2015; 104(6): 1552–60.e1-3

[285]

Mohamed SA, Shalaby SM, Abdelaziz M, Brakta S, Hill WD, Ismail N, Al-Hendy A. Human mesenchymal stem cells partially reverse infertility in chemotherapy-induced ovarian failure. Reprod Sci 2018; 25(1): 51–63

[286]

Abd-Allah SH, Shalaby SM, Pasha HF, El-Shal AS, Raafat N, Shabrawy SM, Awad HA, Amer MG, Gharib MA, El Gendy EA, Raslan AA, El-Kelawy HM. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy 2013; 15(1): 64–75

[287]

Fan D, Wu S, Ye S, Wang W, Guo X, Liu Z. Umbilical cord mesenchyme stem cell local intramuscular injection for treatment of uterine niche: protocol for a prospective, randomized, double-blinded, placebo-controlled clinical trial. Medicine (Baltimore) 2017; 96(44): e8480

[288]

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

[289]

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

[290]

Kilic S, Yuksel B, Pinarli F, Albayrak A, Boztok B, Delibasi T. Effect of stem cell application on Asherman syndrome, an experimental rat model. J Assist Reprod Genet 2014; 31(8): 975–982

[291]

Su J, Ding L, Cheng J, Yang J, Li X, Yan G, Sun H, Dai J, Hu Y. Transplantation of adipose-derived stem cells combined with collagen scaffolds restores ovarian function in a rat model of premature ovarian insufficiency. Hum Reprod 2016; 31(5): 1075–1086

[292]

Terraciano P, Garcez T, Ayres L, Durli I, Baggio M, Kuhl CP, Laurino C, Passos E, Paz AH, Cirne-Lima E. Cell therapy for chemically induced ovarian failure in mice. Stem Cells Int 2014; 2014: 720753

RIGHTS & PERMISSIONS

Higher Education Press

AI Summary AI Mindmap
PDF (3344KB)

5207

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/