Obstet Gynecol Sci Search

CLOSE


Obstet Gynecol Sci > Volume 67(1); 2024 > Article
Chelegahi, Ebrahimi, Reiisi, and Nezamnia: A glance into the roles of microRNAs (exosomal and non-exosomal) in polycystic ovary syndrome

Abstract

Polycystic ovarian syndrome (PCOS) is a common endocrine disorder in women of reproductive age. The clinical symptoms include hyperandrogenism, chronic anovulation, and multiple ovarian cysts. PCOS is strongly associated with obesity and insulin resistance. MicroRNAs (miRNAs) are a group of short non-coding RNAs that play a role in the post-transcriptional regulation of gene expression and translational inhibition. They play a vital role in the regulation of multiple metabolic and hormonal processes as well as in oocyte maturation and folliculogenesis in the female reproductive system. miRNAs can be used as diagnostic biomarkers or therapeutic targets because of their stability. The encapsulation of miRNAs in extracellular vesicles or exosomes contributes to their stability. Exosomes are constantly secreted by many cells and size of about 30 to 150 nm. Enveloping miRNAs exosomes can release them for cellular communication. The induced transfer of miRNAs by exosomes is a novel process of genetic exchange between cells. Many studies have shown that along with non-exosomal miRNAs, different types of exosomal miRNAs derived from the serum and follicular fluid can play an essential role in PCOS pathogenesis. These miRNAs are involved in follicular development and various functions in granulosa cells, apoptosis, cell proliferation, and follicular atresia. The present study aimed to comprehensively review the evidence on miRNAs and their affected pathways under both non-exosomal and exosomal circumstances, primarily focusing on the pathogenesis of PCOS.

Introduction

MicroRNAs (miRNAs) are a class of small, non-coding, and highly conserved RNAs that are ~20-22 nucleotides in length and control many biological functions through the posttranscriptional and translational regulation of mRNA. Binding to the 3′untranslated region (3′UTR) of the target, miRNAs adversely affect their expression, leading to the degradation of target genes’ transcription or translation inhibition [1,2]. miRNAs are transcribed as long primary transcripts from the genome (pri-miRNA) and are then cleaved by the endonuclease enzyme Drosha and the cofactor DGCR8 (DiGeorge syndrome critical region gene 8) in the nucleus [3]. Pre-miRNA, a stem-loop structure of approximately 70-100 nucleotides, is transferred to the cytoplasm by exportin 5 and processed into a double-stranded mature miRNA of approximately 20-22 nucleotides in length through cleavage by Dicer [4,5]. The guide strand of this mature miRNA, which binds to target genes, is associated with the RNA-induced silencing complex (RISC), thus contributing to decreased protein expression [6,7]. This pathway (Drosha/DGCR8) is the canonical pathway for miRNA biosynthesis [8]. In the non-canonical pathway, miRNAs are generated without processing by Drosha/DGCR8 but still require Dicer for maturation, or they are transferred directly from Drosha/DGCR8 to the RISC, and Dicer is not involved [9,10]. miRNAs can regulate multiple target genes simultaneously and are therefore involved in many vital cellular functions, such as the regulation of cell proliferation and differentiation, homeostasis, angiogenesis, development, the immune system, apoptosis, and hormone biosynthesis and secretion [11-15]. miRNA expression is tissue-specific and closely related to tissue function. This feature has also been observed in ovaries [16,17]. The ovary is a complex organ that contains oocytes and somatic cells such as granulosa, theca, and cumulus cells (CCs), which have specific miRNA profiles. Many miRNAs play important roles in the development of follicles, ovulation, and other developmental processes in the ovary [18]. Therefore, miRNAs can be attributed to various types of ovarian disorders and diseases, including polycystic ovary syndrome (PCOS) [19,20]. In patients with PCOS, abnormal expression of miRNAs in serum, follicular fluid, and granulosa cells has been detected, which is reportedly related to anti-apoptotic function and inconclusive testosterone response [17,21].
PCOS is the leading endocrine disorder among women of reproductive age worldwide. The frequency of PCOS is widely dependent on ethnicity, physical characteristics, and diagnostic factors [22,23]. The prevalence of PCOS is calculated as 15% by Rotterdam criteria, reaching 30% in obese and overweight women [24,25]. This disorder is diagnosed based on two of the following three criteria: menstrual irregularity (oligo-ovulation or anovulation), hyperandrogenism (clinical or biochemical), and polycystic ovarian morphology. PCOS is often associated with infertility, obesity, insulin resistance, and dyslipidemia [26-28]. This complex disorder is caused by a combination of environmental and genetic factors [29]. Ethnic diversity in patients with PCOS is strongly associated with their genetic background [30,31]. PCOS is one of the most common endocrine disorders in women and the most common cause of infertility due to lack of ovulation [32]. Moreover, miRNAs play a pivotal role in the posttranscriptional regulation of gene expression and signaling pathways. Therefore, we reviewed recent findings on the expression profiles of exosomal and non-exosomal miRNAs in serum and ovarian follicular fluid and their potential role in ovarian development and PCOS. A better understanding of their functions at different stages of ovarian development can shed new light on women’s reproductive health and the prediction and prevention of various ovarian disorders and infertility.

miRNA and ovarian development

Similar to other human tissues and organs, miRNAs are expressed in ovarian tissue (Fig. 1). These molecules play important roles in ovarian follicular development, including follicular growth, cell proliferation, atresia, and ovulation [33]. Ovarian development begins in the early embryonic period; however, follicular growth and ovulation, followed by luteal tissue development and cell death, continue throughout life. Targeted knockout of argonaute (AGO2) and Dicer in specific tissues of mouse models has revealed the valuable role of miRNAs in ovarian development and the function of various proteins involved in these pathways [34-36]. Bioinformatic and gene ontology analyses have shown that miRNA-related target genes in the ovary are involved in cell cycle regulation, growth, proliferation, apoptosis, endocrine disorders, and ovarian function [37,38]. miRNAs also play a role in the regulation of ovarian follicle growth and are involved in certain stages of ovarian follicular development, ovulation, luteal function, and period.

1. miRNAs in the follicular developmental phase

The miR-17-92 cluster, miR-124, and Let-7 are involved in the growth and development phase of primordial cells; miR-224 in the secondary primordial phase; miR-21, miR-132, miR-212, miR-513a-3p, and miR-125b in the antral phase; and miR-21 and miR-224 in ovulation [18,39]. miR-199a-3p, miR-145, and miR-31 are overexpressed in the follicular phase and substantially decreased during the transition to the luteal-follicular phase [40,41]. miRNAs also play a role in gonadal differentiation. miR-124 is one of the miRNAs that target SOX9 in undifferentiated gonads. Increased expression of SOX9 leads to testicle formation, whereas low levels of SOX9 result in ovarian formation [42]. miR-17-92 cluster miRNAs, except for miR-17-3p, are highly expressed in primordial germ cells (PGCs), and several studies have demonstrated the importance of this cluster in primary ovarian primordial follicle development [43]. For example, miR-143 inhibits primordial follicle formation by reducing the expression of cyclin-dependent kinases (CDKs) 4 and 6 and cyclin B1 in pre-granulosa cells [44]. In mouse ovaries, miR-181a inhibits granulosa cell proliferation by targeting activin receptor IIA [45]. Moreno et al. [46] analyzed the miRNA profiles in the follicular fluid of oocytes in metaphase II (MII), metaphase I (MI), and germinal vesicles (GV). Comparing GV and MII, 13 differentially expressed miRNAs were found, of which miR-451 and miR-563 upregulated in follicular fluid of oocytes in GV and MII stages, respectively. In addition, a comparison of miRNA profiles of the follicular fluid of MII and MI showed that miR-451 was highly expressed in MI and miR-574-5p in MII. Given these findings, they suggested that miR-451 functions as a predictor of the early stages of oocyte maturation. The potential gene targets of these miRNAs are related to the kinase, calcium, and mitogen-activated protein kinase (MAPK) signaling pathways, which are involved in the production of follicle-stimulating hormone and luteinizing hormone (LH) and, in turn, can influence the proper timing of meiosis and oocyte maturation [46]. Studies using mouse models have suggested that the Lin28/Let-7 system is a functional miRNA system involved in ovarian development. Lin28 is an RNA-binding protein that downregulates Let-7. Recent studies have shown that this pathway is involved in the proliferation of PGCs. Although the presence of Let-7 is essential for PGC proliferation and normal mature ovary function during ovarian development and its early stages, the expression of Lin28 is necessary to reduce the expression of Let-7 [47]. A new isoform of Dicer has been identified in recent investigations on mouse and rat oocytes, revealing that Lin28/ Let-7 may play a vital role in oocyte maturation, fertilization, and embryonic development in other species. Let-7b is also expressed in granulosa and CCs of the ovary and is involved in folliculogenesis and ovarian follicle development through regulating the transforming growth factor (TGF)-β signaling pathway [48].

2. miRNAs in the luteal phase

Let-7b, miR -17-5p, miR-122, miR-125, and miR-378 are involved in corpus luteum formation [18,39]. In addition, miR-503, miR-21, and miR-142-3p are generally expressed at lower levels in the follicular stages, and their expression is considerably higher in luteal tissues [40,41]. miRNAs also play a role in the post-ovulation phase, when follicular atresia is caused by granulosa cell apoptosis. MiR-34s activates cell apoptosis and growth arrest through p53 activation and the CDK inhibitor p21 [49]. miR-21 is also highly induced in mouse granulosa cells by corpus LH, and suppression of miR-21 activity in vitro causes granulosa cell apoptosis [50].

Non-exosomal miRNA in PCOS

miRNAs exhibit tissue-specific expression patterns in various organs and systems. Data from various studies have shown that changes in serum levels of miRNAs are valuable and important tools for the diagnosis, tracking, prognosis, and treatment of numerous diseases. This is mostly due to their unique characteristics, such as stability, resistance to nuclease degradation, simple detection, and the potential to be disease-specific [51]. The mechanism by which miR-NAs are transferred from inside the cell to the extracellular environment, such as serum, is accomplished through two main pathways: 1) active transfer via extracellular vesicles, such as exosomes and 2) transfer as part of a protein-miRNA complex; they can also leak from damaged cells [52]. Table 1 shows the various miRNAs and their potential functions in different parts of ovarian tissue.

1. Serum

Several studies have shown that changes occur in the serum levels of different miRNAs in patients with PCOS (Fig. 2). Since serum levels are easily detectable, they can be used as important non-invasive biomarkers for early diagnosis of PCOS [53]. Long et al. [54] observed that increased serum levels of miR-222, miR-146a, and miR-30 might play a role in the pathogenesis of PCOS. Bioinformatics analysis showed that the target genes of these miRNAs were involved in metastasis, cell cycle, apoptosis, and endocrine pathways [54]. In patients with PCOS, the level of testosterone increases, and insulin contributes to this by upregulating the synthesis and accumulation of androgens in the adipose tissue [55]. In addition, miR-132, miR-320, miR-520c-3p, miR-24, and miR-222 regulate estradiol concentrations, while miR-24, miR-193b, and miR-483-5p regulate progesterone concentrations in patients with PCOS [56]. Studies have demonstrated that miR-222 is directly correlated with serum insulin levels, and miR-146a is inversely associated with serum testosterone [57]. Changes in the serum levels of miR-222, miR-146a, and miR-30 have been introduced as new biomarkers for the diagnosis [42]. miRNAs are also involved in the pathogenesis of PCOS through metabolic processes. For example, miR-93 is overexpressed in PCOS and associated with decreased glucose transporter 4 (GLUT4) expression and increased insulin resistance [58]. Additionally, miRNA-21, miRNA-27b, miRNA-103, and miRNA-155 play important roles in metabolic processes and are affected by obesity and circulating androgen concentrations in patients with PCOS [59]. miR-103 is one of the miRNAs involved in insulin resistance and obesity and plays a role in regulating testosterone levels and the occurrence of PCOS. Additionally, it increases in serum and is therefore easily detectable; making its serum level be considered as a biomarker for PCOS, especially in obese women [60]. Research conducted on miR-23a and miR-23b showed a positive effect of obesity on the serum expression of miR-23b, an adverse effect of testosterone on the expression of miR-23a, and decreased expression of these two miR-NAs in the serum of patients with PCOS compared to healthy women. Although, miR-23a is a more effective option concerning new biomarkers for PCOS diagnosis [61]. miRNA-93 and miRNA-223 were significantly upregulated in the blood of patients with PCOS compared to controls. There is no association between miRNA-93 or miRNA-223 with insulin, homeostatic model assessment for insulin resistance, homeostasis model assessment of beta cell function, or testosterone levels in both groups. Plasma levels of miR-93 can differentiate patients with PCOS from healthy subjects with greater sensitivity and specificity than miR-223. Thus, circulating miR-93 could serve as a biomarker for PCOS [62]. miR-320 is one of the other serum miRNAs related to the PCOS phenotype, whose serum level decreases compared to the normal group, and can also be used as a non-invasive diagnostic biomarker [63,64]. In addition, miR-320 is downregulated in follicular fluid [56] and ovarian tissue [65]. Endothelin (ET-1) is a bioactive peptide produced by endothelial cells that participates in tumor growth by enhancing cell mitosis [66]. miR-320 is involved in follicular maturation by targeting the ET-1 gene, and a decrease in miR-320 can lead to the progression of the PCOS phenotype in affected women [46]. Changes in miR-NA-21 expression were observed in both ovarian tissue and serum. Increased expression of miR-21 acts as an inhibitor of the serine/threonine protein kinase LATS1 and stimulates the abnormal growth of secondary follicles by affecting the Hippo signaling pathway and increasing the level of cellular communication network protein, which is involved in the further growth, viability, and proliferation of cells [67]. miRNA-6767-5p, involved in the cell cycle and immune system processes, plays an important role in ovarian development [49]. In a study conducted on Korean women with PCOS, Sang et al. [56] observed that the serum level of miR-6767-5p was inversely correlated with fasting glucose levels and directly correlated with the number of menstrual periods per year. As hyperandrogenemia is the main characteristic of PCOS, miR-6767-5p may be of great importance in the pathogenesis and metabolic manifestations of PCOS. They suggested that serum miR-6767-5p may serve as a new molecular marker for the diagnosis of PCOS in Korean women [68].

2. Ovarian tissue

In a study of the expression profiles of miRNAs in the ovarian tissue of women with PCOS, Baley and Li [35] observed that some miRNAs, such as miR-146a, miR-22, miR-132, miR-200c, miR-141, and miR-21, showed significantly increased expression. Interestingly, most differentially expressed miRNAs in the ovarian tissue of patients with PCOS were expressed at normal levels in serum. Therefore, all miRNAs in the PCOS ovarian tissue are released into the blood. MiR-142, miR-33b, and miR-423 are other miRNAs involved in PCOS that contribute to the initiation and progression of the PCOS phenotype by targeting Is transforming growth factor beta receptor 1 and mothers against decapentaplegic homolog 7 (SMAD7) and inhibiting the TGFB signaling pathway [69]. Bioinformatics analyses have revealed that dysregulation of miR-15b and miR-497, which are miRNAs involved in the insulin signaling pathway, contributes to PCOS pathogenesis [70]. miR-320 also regulates the expression of insulin receptor substrate (IRS)-1 and the extracellular signal-regulated kinase (ERK) 1/2 activity in ovarian tissue, which are related to insulin resistance in PCOS. Therefore, understanding the mechanism of action of miRNAs in PCOS patients with insulin resistance presents a theoretical basis for the diagnosis and treatment of this condition [65,71]. The inhibition of miR-483 expression has been observed in the ovarian cortex of patients with PCOS. miR-483 is downregulated in PCOS and prevents granulosa cell proliferation by targeting insulinlike growth factor (IGF)-1 [72]. miR-33b-5p is overexpressed in the ovarian tissues of insulin-resistant mice with PCOS and thus may play an important role in the development of insulin resistance in patients with PCOS. Additionally, miR-33b-p inhibited the production of GLUT4 by targeting high-mobility group AT-hook 2 [73]. Another study identified 29 differentially expressed miRNAs in patients with PCOS. In these patients, miR-382-5p correlated with age and free androgen index (FAI), miR-199b-5p with anti-müllerian hormone (AMH), and miR-93-3p with C-reactive protein. In healthy controls, miR-127-3p, miR-382-5p, and miR-425-3p correlate with fertilization rate, miR-127-3p with insulin resistance, and miR-381-3p with FAI [74].

3. Follicular fluid

Ovarian follicular fluid plays an important role throughout follicular development. Being a complex mixture of hormones, growth factors, anti-apoptotic factors, proteins, and miRNAs secreted from the granulosa and theca cells, it provides a vital microenvironment for oocyte development and maturation of oocytes [75]. Scalici et al. [76] reported that the expression of Let-7b and miR-140 is significantly decreased, whereas the expression of miR-30a is increased in the follicular fluid of patients with PCOS. The results showed that reduced expression of Let-7b and miR-140 contributed to the progression of the PCOS phenotype. In addition, the change in the expression of estrogen receptor in PCOS was attributed to reduced expression of miR-140 in the follicular fluid [76].
In Roth et al. [77] study, five overexpressed miRNAs (hsamiR-9, 18b, 32, 34c, and 135a) were found in follicular fluid, and three of their potential targets were associated with the PCOS phenotype (IRS-2, synaptotagmin 1, and interleukin 8). These three genes are involved in carbohydrate metabolism, beta cell function, cell-cell communication, and steroid synthesis in the late follicular phase [77]. Some miRNAs are involved in the metabolism of hormones; for example, changes in the expression of some miRNAs involved in the metabolism of androgens have been observed in the follicular fluid of patients with PCOS. Among these, miR-151 was inversely correlated with free testosterone, whereas miR-29a, miR-320, and miR-518 were directly correlated with testosterone levels. miR-132, miR-135, and miR-146a inhibit progesterone and testosterone secretion, whereas miR-9, miR-18b, and miR-155 inhibit testosterone secretion [75]. In this regard, Naji et al. [78] observed that the intermediary role of (miR-93 and miR-21) miRNAs in granulosa cells and follicular fluid plays an important role as an androgen-responsive factor in the pathogenesis of PCOS in hyperandrogenism.

4. Granulosa cells

miR-145 plays a prominent role in granulosa cell proliferation, follicle selection, and folliculogenesis. And miR-182 plays an essential role in the appropriate functioning of granulosa cells by regulating proliferation, apoptosis, and steroidogenesis. Thus, aberrant expression of miR-182 (in follicular fluid) and miR-145 (in granulosa cells) may contribute substantially to follicular developmental disorders in PCOS [78]. Overexpression of miR-145 inhibits the mitogenactivated protein kinases (MAKP)/ERK signaling pathway in granulosa cells. These findings have also shown that miR-145 can suppress cell proliferation and that the mechanism involved is related to the suppression of the expression of IRS-1, which leads to the inhibition of the MAKP/ERK signaling pathways. In addition, high insulin concentrations decreased miR-145 expression, regulated IRS-1 protein expression, and promoted cell proliferation [79]. The expression of miR-483-5p in PCOS granulosa cells also increases and regulates the MAPK3 pathway [80]. Dihydrotestosterone (DHT) increases granulosa cell apoptosis and, at high concentrations, inhibits miR-182 expression. miR-182 inhibits granulosa cell apoptosis by targeting SMAD7, as well [81]. miRNAs are involved in the regulation of various pathways, biological functions, and cellular components in PCOS. Xue et al. [82] reported increased expression of miR27a-3p in PCOS mouse ovaries, and its function was investigated in mouse primary granulosa cells. They confirmed that the expression of miR-27a-3p in mouse granulosa cells increases in the pre-antral follicle stage and that the cyclic AMP response element binding protein 1 is a target gene for miR-27a-3p, which prevents the expression of the downstream factor CYP19A1 (cytochrome P450 family 19 subfamily A member 1). The imbalance in androgen and estradiol levels under the influence of miR-27a-3p and its function in promoting granulosa cell apoptosis may be involved in the pathophysiology of PCOS [83]. In addition, Zhang et al. [84] observed that a reduction in miR-320a impairs steroidogenesis in cumulus granulosa cells by dysregulating the function of the bone transcription factor RUNX2. CCs are the main site of estrogen synthesis. Bidirectional communication between CCs and follicular fluid is essential for ovarian steroidogenesis. miR-320a is markedly decreased in the primary CCs of patients with PCOS, which results in a sharp drop in estrogen levels in CCs. IGF-1 regulates the expression of miR-320a in CCs, and miR-320a can promote steroidogenesis in CCs by decreasing the expression of CYP11A1 and CYP19A1 by directly targeting the 3’UTR of RUNX. Therefore, deregulation of the miR-320a/RUNX2/CYP11A1 (CYP19A1) cascade plays an important role in the development of estrogen deficiency in human CCs. Androgens influence the growth, health, and survival of follicles and the pathophysiology of PCOS. Investigations have revealed that miR-93 and miR-21 are increased in the granulosa cells of hyperandrogenic PCOS patients compared to normoandrogenic patients. Free testosterone and FAI were directly correlated with miR-93 and miR-21 in PCOS Granulosa cells. Thus, as miR-93 and miR-21 are widely known androgen-responsive factors, they may be involved in follicular dysfunction during the pathogenesis of PCOS in hyperandrogenism [85]. Transfecting miR-509-3p mimic in KGN cells and studying E2 production confirmed that miR-509-3p improves estradiol secretion by inhibiting MAP3K8 expression. These results help to define the pathogenesis of anovulation in PCOS, particularly the regulation of estradiol secretion [86]. miR-93 expression is upregulated in granulosa cells of PCOS patients and inhibits cyclin-dependent kinase inhibitor 1A (CDKN1A) in PCOS granulosa cells. MiR-93 causes granulosa cell proliferation and G1/S transition. Therefore, miR-93 promotes ovarian granulosa cell proliferation through targeting CDKN1A in PCOS [21,87].

5. Theca cells

Ovarian theca cells play a vital role in ovarian follicular development and hormone secretion [88]. Differential expression of some miRNAs has been reported in theca cells, which may regulate various signaling pathways including meiosis, WNT, TGF-β, and MAPK [89]. A study of miRNAs in atretic follicles compared to healthy cattle follicles revealed that miR-150, miR-409a, miR-142-5p, miR-222, miR-155, and miR-199a-5p were more highly expressed in the theca than in the granulosa cells of atretic follicles [90]. In a rat POCS model induced by DHT, most of the miRNAs that stimulated cyst formation were expressed in follicular theca cells [91,92]. Hossain et al. [93] reported the upregulation of miR-222 in theca cells of PCOS samples; however, androgens can target P27/kip1(a cyclin-CDK inhibitor) to regulate the proliferation of theca cells, leading to miR-222 suppression. In vitro studies have confirmed that PCOS theca cells secrete higher androgen levels than normal cells [94]. In addition, miR-103a-3p and miR-376a-3p positively correlated with androgen levels in PCOS [95]. Therefore, miRNAs may be implicated in pathways involved in insulin signaling, steroid biosynthesis, endothelial regulation, and folliculogenesis.

Exosomal miRNA in PCOS

Exosomes are a heterogeneous group of extracellular microspheres with diameters of approximately 4-100 nm [96]. Exosomes exist in different types of mammalian cells in the form of multivesicular bodies in the intracellular space and are secreted out of the cell or into biological fluids such as joint fluid, amniotic fluid, plasma, cerebrospinal fluid, urine, saliva, breast milk, alveolar fluid, follicular fluid, and bile through integration with the plasma membrane [97,98]. Exosomes may have different surface markers depending on their cellular origin and physiological state. They contain a variety of cell/tissue-specific proteins, RNAs, lipids, and biological biomolecules that facilitate cell communication with the surrounding environment and have a significant impact on biological and pathological events [99]. AGO2 and some RNA-binding proteins such as hnRNPA2B [100] and Y-box1 [101] are involved in the regulation of miRNA loading into exosomes. During the formation of multivesicular bodies in the cytoplasm, AGO2 binds to the exosome marker protein CD63, leading to the loading of miRNAs into exosomes [102]. Table 2 displays the exosomal miRNAs compared to non-exosomal miRNAs in patients with PCOS.

1. Serum

Che et al. [103] exposed endometrial cancer cell lines to serum exosomes isolated from patients with PCOS and found that the rates of migration and invasion of cancer cells increased in vitro. Further investigations showed that levels of miR-27-5p in the serum exosomes of patients with PCOS were significantly increased. They observed that exosomal miR-27-5p stimulated the migration and invasion of endometrial cells by targeting SMAD4 and reducing its expression. Therefore, miR-27-5p plays an important role in the esophageal carcinoma progression of endometrial cancer in PCOS patients [103]. Furthermore, the interaction between insulin resistance and obesity in PCOS may be related to the regulation of exosome-secreted miRNAs. An insulin resistance-based model of PCOS was established in rats with dehydroepiandrosterone (DHEA) on a high-fat diet (HFD). Expression of miR-20b-5p and miR-106a-5p was detected in serum exosomes. Overexpression of serum exosomal miR-20b-5p and miR-106a-5p inhibits adipocyte differentiation in 3T3-L1 cells in a mouse model of PCOS with IR. In DHEA+HFD serum-derived exosomes, miR-20b-5p and miR-106a-5p levels were significantly decreased. Overexpression of miR-20b-5p and miR-106a-5p decreased the expression of genes related to adipose differentiation and triglyceride content in 3T3-L1 cells of mice with liver steatosis. Serum-derived exosomal miR-20b-5p and miR-106a-5p inhibit adipocyte differentiation during PCOS with IR and therefore have the potential to serve as new therapeutic targets [104]. Another study has investigated the expression profiles of exosomal miRNAs in PCOS and non-PCOS patients. Hsa-miR-1299, hsa-miR-145-5p, hsa-miR-6818-5p, and hsa-miR-192-5p were found to be differentially expressed in the serum exosomes of PCOS patients and therefore have the potential to be used as diagnostic biomarkers for PCOS [105]. Circulating exosomes in the PCOS plasma contain differentially expressed miRNAs. In patients with PCOS, the expression levels of miR-146a-5p and miR-126-3p increased, whereas those of miR-20b-5p, miR-106a-5p, and miR-18a-3p decreased. The differential expression of these miRNAs has been suggested to target various functions, including the MAPK signaling pathway, axon guidance, endocytosis, and tumorigenic pathways. Therefore, these exosomal miRNAs may be associated with PCOS [106].

2. Follicular fluid and other cells

Various studies have shown that more than 200 different miRNAs exist either freely or inside human follicular fluid exosomes [107]. Fig. 3 shows some miRNAs in the follicular fluid exosomes. Developmental pathway analysis showed that the exosomal and non-exosomal miRNAs target genes are involved in MAPK, Wnt, FOXO, nuclear factor kappa B, Oxytocin, epidermal growth factor (ErbB), PI3K-Akt, and neurotrophin signaling pathways, which play role in follicular development, cell proliferation, and resumption of meiosis [108]. Li et al. [109] study showed that the S100-A9 protein in exosomes could activate NF-κB signaling pathways in granulosa cells, increase the production of inflammatory cytokines, and disrupt steroidogenesis. These findings indicate the importance of exosomes as extracellular mediators in the pathophysiology of PCOS [109]. Increased expression of miR-29a, miR-99a, miR-100, miR-132, miR-212, miR-214, miR-218, miR-508-3p, and miR-654-3p, all molecular regulators of ovarian follicle development, was observed in exosomes isolated from the follicular fluid. Some of these miRNAs or their host genes are expressed in granulosa or CCs. However, they also play a role in targeting genes within follicular cells. Most of the upregulated miRNAs are involved in the regulation of WNT, MAPK, ErbB, and TGF-β signaling pathways, and can contribute to the development of ovarian follicles or stimulate the proliferation of granulosa cells and expansion of cumulus. They can also start the resumption of meiosis by negatively regulating genes that encode follicle maturation-inhibiting factors [110]. Hu et al. [111] identified six exosomal miRNAs in the follicular fluid (miR-6087, miR-199a-5p, miR143-3p, miR-483-5p, miR-23b-3p, and miR-200a-3p) related to PCOS pathogenesis and predicted that these miRNAs were mainly related to amino acid biosynthesis; metabolism of glycine, serine, and threonine; glycosaminoglycan biosynthesis; monocarboxylic acid metabolism; and carbon metabolism. Yang et al. [112] introduced three exosomal miRs, miR-125b, miR-19b, and miR-222, found in the follicular fluid associated with PCOS pathogenesis.
Inhibition of apoptosis and increased senescence in granulosa cells can also be mediated by exosomal miRNAs. The expression of miR-424-5p significantly decreases in PCOS exosomes and primary granulosa cells. Exosomes enriched with miR-424-5p significantly increased granulosa cell senescence and suppressed cell proliferation. Consistent with the results obtained in cells transfected with miR-424-5p mimic, miR-424-5p caused senescence and inhibited cell proliferation. The cell division cycle 4 (CDCA4) was identified as a direct target of miR-424-5p. Overexpression of CDCA4 reverses the effects of exosomal miR-424-5p on granulosa cells, activates the Rb/E2F1 signaling pathway, induces cellular senescence in PCOS, and attenuates the disease [113]. However, exosomal miRNAs can induce opposite effects. For example, exosomes containing miR-143-3p derived from PCOS follicular fluid increased granulosa cell apoptosis by targeting bone morphogenetic protein receptor and blocking the Smad1/5/8 signaling pathway [114]. miR-18b-5p produced by follicular fluid-derived exosomes decreases phosphatase and tensin homolog expression and activates the the phosphatidylinositol-3-kinase/akt and the mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway to improve PCOS [115].

Significance of miRNA in therapy

miRNAs are among the most significant RNA groups in clinical research. Because a specific miRNA may be involved in different biological pathways disrupted in a disease, it can be very useful from a therapeutic perspective, especially if the disease is not associated with a genetic defect. In this regard, bioinformatics software used to identify miRNA binding sites, target genes, and the biological pathways involved, along with laboratory in vitro and in vivo research models, have greatly assisted in accelerating the identification of miR-NAs in clinical medicine [116]. In healthy individuals, a large proportion of ovarian follicles undergo apoptosis and die during their reproductive life through a vital process known as follicular atresia [117]. Follicular atresia begins with the apoptosis of follicular granulosa cells (granulosa cells), followed by the degeneration of other follicular components. Factors such as IGF-1 are follicle viability factors that control atresia [118]. Das et al. [119] showed that there was a significant variation in the apoptosis rate and proliferation of granulosa cells in patients with PCOS. They were the first to find that the level of active caspase-3, which is functionally required to initiate cell death, was significantly lower in the granulosa cells of anovulatory PCOS follicles than in those of normal ovulatory follicles. The expression of Bcl-XLong, an anti-apoptotic factor, increased approximately two-fold, while that of Bax, an apoptosis inducer, slightly decreased in the PCOS group. The expression of Ki-67 (an indicator of cell proliferation) was significantly higher in the PCOS group, indicating a significant imbalance between the rates of apoptosis and proliferation in patients with PCOS [119]. Studies have shown that miRNAs are involved in this pathway, and their regulation can be useful in disease treatment. miR-99a is one of the miRNAs found in granulosa cells, and its expression is significantly decreased in PCOS. This decrease was associated with an increase in the IGF-1R protein levels. IGF-1R belongs to the transmembrane receptor tyrosine kinase family and is activated by IGF-1 [120]. During follicular development, the increase in IGF-1R through the stimulation of tyrosine kinase pathways leads to an increase in proliferation and a decrease in apoptosis of granulosa cells in the first stage of folliculogenesis, which contributes to the pathogenesis of PCOS. Therefore, the re-regulation of miR-99a may be an effective method for reducing the adverse effects of IGF-1/IGF-1R system overactivation in PCOS [121]. miR-323-3p is another miRNA involved in the pathogenesis of PCOS that regulates cell apoptosis and steroidogenesis by targeting IGF-1. Studies have shown that the expression of miR-323 declines in CCs of PCOS patients [122]. CCs, a subclass of granulosa cells that surround the oocyte in the antral follicle, are the source of ovarian steroid metabolism and growth hormones and play an important role in oocyte maturation [123]. Apoptosis is one of the primary causes of PCOS [124]. Increased miR-323-3p expression in CCs stimulates cell proliferation and inhibits apoptosis. As previously mentioned, CYP19A is a critical enzyme in the production of ovarian estradiol, which increases in CCs in PCOS [125]. The expression level of this enzyme is indirectly controlled by miR-323; thus, the reduction of miR-323 in PCOS CCs leads to an increase in the level of IGF-1, followed by an increase in CYP19A, and a disruption in the completion of oocyte development. Thus, miR-323-3p is a novel and promising molecular target for ameliorating CC dysfunction in PCOS [105]. Programmed cell death 4 (PDCD4) is a key factor in apoptosis that suppresses protein translation by binding to eIF4A and inhibiting RNA helicase activity [126]. Available evidence demonstrates that the expression of PDCD4 increases in patients with PCOS and is involved in the pathogenesis of the disease [127]. In an investigation of 36 female C57BL/6 mice, Zhao et al. [128] observed high PDCD4 expression in mice with PCOS compared to that in controls. Delivery of exogenous miR-323-3p by adipose mesenchymal stem cell derived exosomes can suppress the expression of PDCD4. As the apoptosis of CCs increases in PCOS to induce premature follicular atresia, studies have suggested that PDCD4 contributes to premature apoptosis in PCOS CCs, and its pro-apoptotic effect can be eliminated by miR-323-3p. Therefore, miR-323-3p upregulation suppresses CCs apoptosis by targeting PDCD4 in PCOS, and the delivery of miR-323-3p via adipose-derived mesenchymal stem cell exosomes is an effective approach for regulating PCOS progression [128]. miR-135 is another miRNA found in granulosa cells and its expression is highly upregulated in PCOS. miR-135a is regulated by androgens at the transcriptional level. Increased androgen levels in PCOS lead to an increase in miR-135a, and miR-135a can suppress proliferation, enhance the DNA damage response, and induce apoptosis by directly binding to the vascular endothelial growth factor C gene. Consequently, the rate of apoptosis increases during the middle stage of the antrum, leading to cyst formation. Therefore, regulating miR-135a expression can be considered a new therapeutic goal for PCOS patients [129]. miR-335-5p expression is inversely correlated with the number of antral follicles, AMH and testosterone levels. Research has shown that miR-335 levels in the granulosa cells and follicular fluid are significantly decreased in patients with PCOS. Bioinformatics analyses have shown that SGK3 is the direct target of this miRNA, a member of the family of kinases involved in cell proliferation through the PI3K/AKT-mTOR signaling pathway [130,131]. In granulosa cells, SGK is localized in the cell nucleus and regulates cell proliferation and differentiation. In the early stages of folliculogenesis, a reduction in miR-335 levels leads to an increase in AMH levels and granulosa cell proliferation. AMHs prevent the premature depletion of ovarian follicles and regulate steroidogenesis in granulosa cells [132]. In contrast, the reduction of miR-335 expression in an SGK-dependent pathway leads to increased proliferation of granulosa cells, and delayed follicular atresia, and contributes to PCOS pathogenesis. These observations provide a new perspective on the function of granulosa cells in PCOS and suggest that miR-335-5p can serve as a new molecular target for improving dysfunctional granulosa cells [131]. Díaz et al. [133] investigated circulatory concentrations of miR-451a, miR-652-miR-451a, miR-652-3p, miR-106b-5p, and miR-206 in girls with PCOS treated with oral contraceptive or spironolactone-pioglitazone-metformin. They observed that the concentration of the studied miRNAs decreased significantly compared with that in the control group. These target genes are involved in energy homeostasis and cell-cycle regulation. This study demonstrated the capability of miR-451a to diagnose PCOS with 100% sensitivity [133].
Additionally, alterations in the circadian rhythm can trigger PCOS through oxidative stress and inflammation. Circadian rhythms can alter circulating exosomes and related miRNAs, leading to alterations in physiological conditions [134]. miR-NAs play an important role in maintaining homeostasis of the circadian system, and miR-132 and miR-219 are involved in regulating circadian rhythms in mammals by targeting clock genes. Furthermore, miR-494, miR-27b-3p, miR-155, and miR-142-3p are involved in regulating the clock gene, brain, and muscle Arnt-like protein 1 (BAML1) [135,136]. Changes in the expression of miR-192 and miR-194 not only affect the rhythm fluctuation of BMAL1 mRNA but also suppress the expression of the clock gene period (Per) [137]. Therefore, circadian rhythms can be modulated by targeting miRNAs to investigate their potential in PCOS treatment [134].

Conclusion

miRNAs that are free or enclosed in extracellular vesicles (exosomes) can affect various signaling pathways. Abnormal expression of miRNAs occurs in the granulosa cells, theca cells, adipose tissue, follicular fluid, serum, and peripheral blood leukocytes of women with PCOS. They regulate follicular growth and maturation, insulin signaling, glucose and lipid metabolism, and steroid hormone synthesis. Additionally, exosomes can protect miRNAs from degradation, making them stable and efficient diagnostic tools.
The use of miRNAs in the clinical management of PCOS is a dynamic part of research, and there are several potential functions of miRNAs in this context. miRNAs can be used clinically in various pathways, such as diagnostic and prognostic biomarkers, therapeutic targets, and drug delivery vehicles; therefore, as diagnostic and prognostic biomarkers of PCOS. Many studies have identified specific miRNAs that are dysregulated in individuals with PCOS, and their detection in the blood or other fluids may be useful for early diagnosis and monitoring of disease progression. Also, the expression of certain miRNAs has been found to be associated with insulin resistance, a common feature of PCOS. By measuring the levels of these miRNAs, it may be possible to predict the probability of increased IR-related complications, such as type 2 diabetes.
Although miRNAs are potentially promising diagnostic and therapeutic factors, their clinical applications in diagnosis and treatment are still under development. Concerning diagnosis, many of the identified miRNAs are still at the experimental level and require further investigation. Additionally, only a limited number of miRNAs have been approved by the Food and Drug Administration as therapeutic agents. Therefore, the application of miRNAs as diagnostic or therapeutic factors in medicine is in its early stages, and there is still a long way to go.

Notes

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

This article does not contain any studies on human or animals.

Patient consent

This article does not contain any individual person’s data in any form.

Funding information

No fund.

Fig. 1
MicroRNAs (miRNAs) involved in the regulation of ovarian development.
ogs-23193f1.jpg
Fig. 2
Non-exosomal microRNA (miRNA) in polycystic ovarian syndrome (PCO). Red arrow represents upregulated miRNA, blue arrow represents down-regulated miRNA [56].
ogs-23193f2.jpg
Fig. 3
MicroRNAs (miRNAs) in follicular fluid exosomes and their functions. Red DNA represents upregulated miRNAs and blue DNA represents downregulated miRNAs in exosomes. WNT, wingless-related integration site; MAPK, mitogen-activated protein kinases; TGF, transforming growth factor; ErbB, human epidermal growth factor receptor.
ogs-23193f3.jpg
Table 1
Types of miRNAs and their target genes and functions in different parts of the ovary
miRNA Tissue/cell Target gene Expression Function
miR-21 Follicle Serine/threonine protein kinase LATS1, effects on the Hippo signaling pathway Up Stimulator of abnormal secondary follicle growth
miR-34s Granulosa cells p53 and the cyclin-dependent kinase inhibitor p21 Up Activation of cell apoptosis and growth arrest
miRNA-124 Undifferentiated gonads sox9 Down Ovary formation
Let-7b Granulosa cells and cumulus cells TGF-β signaling pathway Up Folliculogenesis and follicle development
miR-483 Ovarian cortex IGF-1 Down Inhibits the proliferation of granulosa cells
miR-103 Serum Insulin resistance pathway and obesity Up Changes in testosterone levels and the occurrence of PCOS
miR-320a Granulosa cells and cumulus cells, serum ET-1 Down Disruption of steroidogenesis, diagnostic biomarker
miR-93 Granulosa cells GLUT4 Up Proliferation of granulosa cells and transition from G1 to S
miR-182 Granulosa cells Smad7 Up Inhibition of granulosa cell apoptosis
miR27a-3p/5p Granulosa cells, serum, KK-1 SMAD4 Up Promotes apoptosis and inhibits estradiol (E2) production in KK-1 cells

miRNA, microRNA; LATS1, large tumor suppressor kinase 1; TGF, transforming growth factor; IGF, insulin-like growth factor; PCOS, polycystic ovarian syndrome; ET-1, endothelin 1; GLUT4, glucose transporter 4; KK-1, immortalized murine ovarian granular cells; SMAD7, mothers against decapentaplegic homolog 7.

Table 2
Non-exosomal miRNAs and their functions compared with exosomal miRNAs in PCOS
miRNA Site Target gene Status Function
Non-exosomal
 miR-222, miR-146a and miR-30, miR-23a, miR-23b Serum Target genes in metastasis, cell cycle, apoptosis and endocrine pathways Down Pathogenesis of PCOS
 miR-520c-3p, miR-24 and miR-222 Serum - Up Regulation of estradiol concentration
 miR-24, miR-193b, miR-483-5p Serum - Up Regulation of progesterone level
 miR-93, miRNA-223 Serum GLUT4 Up Increased insulin resistance, Biomarkers in PCOS
 miR-103 Serum - Up Insulin resistance and obesity
 miR-132, miR-320 Serum, ovarian tissue, follicular fluid - Down Regulation of IRS-1 and ERK1/2 activity
 miRNA-21 Serum, ovarian tissue, granulosa cell, follicular fluid - Up Abnormal growth of the secondary follicle
 miRNA-6767-5p Ovarian tissue - Down Mainly involved in cell cycle and immune system processes, ovarian growth
 miR-142, miR-423 Ovarian tissue TGFBR1 and SMAD7 Up Progression of the PCOS phenotype
 miR-483 Ovarian tissue IGF-1 Down Prevents GC proliferation
 Let-7b, miR-140 Follicular fluid - Down Progression of the PCOS phenotype
 miR-30a, miR-145 Follicular fluid - Up Progression of the PCOS phenotype/ proliferation of GC cells
 miR-182 Follicular fluid SMAD7 Up Regulation of proliferation, apoptosis and steroidogenesis
 miRNA-483-5p Granulosa cells - Up MAPK3 pathway regulation
 miR-27a-3p Follicular fluid Creb1 gene Up Inhibition of CYP19A1 expression
 miR-320a Granulosa cells - Down Strong reduction of estrogen in CCs
 miR-509-3p - MAP3K8 gene Down Improved secretion of E2
 miR-93 Granulosa cells, follicular fluid CDKN1A gene Up Increased ovarian GC proliferation
 miR-33b-p Granulosa cells HMGA2 gene Up Inhibition of GLUT4 production
Exosomal
 miR-424-5p Granulosa cells CDCA4 gene Down Suppression of cell proliferation
 miR-143-3p Granulosa cells BMPR1A gene Up Blocking the Smad1/5/8 signaling pathway
 miR-18b-5p Follicular fluid PTEN gene Up Activation of the PI3K/Akt/mTOR signaling pathway
 miR-29a, miR-99a, miR-100, miR-132, miR-212, miR-214, miR-218, miR-508-3p, miR-654-3p Follicular fluid - Up Molecular regulators of ovarian follicle growth and development pathways
 miR-6087, miR-199a-5p, miR143-3p Follicular fluid Up Pathways related to amino acid biosynthesis, monocarboxylic acid metabolism and carbon metabolism
 483-5p, miR-23b-3p, miR-200a-3p Follicular fluid - Down Amino acid and carbon metabolism
 miR-125b, miR-19b, miR-222 Follicular fluid - Up Associated with the pathogenesis of PCOS
 miR-27-5p Serum SMAD4 gene Up Progression of endometrial cancer
 miR-20b-5p, miR-106a-5p Serum Genes related to fat differentiation Down Inhibition of fat cell differentiation
 miR-1299, miR-6818-5p Serum - UP Biomarkers in the diagnosis of PCOS
 miR-145-5p, miR-192-5p Serum - Down Biomarkers in the diagnosis of PCOS

miRNA, microRNA; PCOS, polycystic ovarian syndrome; GLUT4, glucose transporter 4; IRS, insulin receptor substrate; ERK, extracellular signal-regulated kinase; TGFBR1, transforming growth factor beta receptor 1; SMAD7, mothers against decapentaplegic homolog 7; IGF, insulin-like growth factor; GC, granulosa cell; MAPK, mitogen-activated protein kinase; Creb1, CAMP responsive element binding protein 1; CYP19A1, cytochrome P450 family 19 subfamily A member 1; CCs, cumulus cells; CDKN1A, cyclin-dependent kinase inhibitor 1A; HMGA2, high-mobility group AT-hook 2; CDCA4, cell division cycle 4; BMPR1A, bone morphogenetic protein receptor, type 1A; PTEN, phosphatase and tensin homolog; PI3K/Akt/mTOR, the phosphatidylinositol-3-kinase/akt and the mammalian target of rapamycin; CAMP, cyclic adenosine monophosphate.

References

1. Ambros V. The functions of animal microRNAs. Nature 2004;431:350-5.
crossref pmid pdf
2. Vishnoi A, Rani S. MiRNA biogenesis and regulation of diseases: an overview. Methods Mol Biol 2017;1509:1-10.
crossref pmid
3. Lee Y, Ahn C, Han J, Choi H, Kim J, Yim J, et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003;425:415-9.
crossref pmid pdf
4. Lund E, Güttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science 2004;303:95-8.
crossref pmid
5. Kirstein N, Dokaneheifard S, Cingaram PR, Valencia MG, Beckedorff F, Gomes Dos Santos H, et al. The integrator complex regulates microRNA abundance through RISC loading. Sci Adv 2023;9:eadf0597.
crossref pmid pmc
6. Martinez J, Patkaniowska A, Urlaub H, Lührmann R, Tuschl T. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell 2002;110:563-74.
crossref pmid
7. Schwarz DS, Hutvágner G, Haley B, Zamore PD. Evidence that siRNAs function as guides, not primers, in the Drosophila and human RNAi pathways. Mol Cell 2002;10:537-48.
crossref pmid
8. Michlewski G, Cáceres JF. Post-transcriptional control of miRNA biogenesis. RNA 2019;25:1-16.
crossref pmid pmc
9. Castellano L, Stebbing J. Deep sequencing of small RNAs identifies canonical and non-canonical miRNA and endogenous siRNAs in mammalian somatic tissues. Nucleic Acids Res 2013;41:3339-51.
crossref pmid pmc
10. Achkar NP, Cambiagno DA, Manavella PA. miRNA biogenesis: a dynamic pathway. Trends Plant Sci 2016;21:1034-44.
crossref pmid
11. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH, et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 2007;26:745-52.
crossref pmid pmc
12. Baltimore D, Boldin MP, O’Connell RM, Rao DS, Taganov KD. MicroRNAs: new regulators of immune cell development and function. Nat Immunol 2008;9:839-45.
crossref pmid pdf
13. Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci U S A 2008;105:5166-71.
crossref pmid pmc
14. Xiao C, Rajewsky K. MicroRNA control in the immune system: basic principles. Cell 2009;136:26-36.
crossref pmid
15. Taheri F, Ebrahimi SO, Shareef S, Reiisi S. Regulatory and immunomodulatory role of miR-34a in T cell immunity. Life Sci 2020;262:118209.
crossref pmid
16. Li Y, Fang Y, Liu Y, Yang X. MicroRNAs in ovarian function and disorders. J Ovarian Res 2015;8:51.
crossref pmid pmc pdf
17. Chen B, Xu P, Wang J, Zhang C. The role of MiRNA in polycystic ovary syndrome (PCOS). Gene 2019;706:91-6.
crossref pmid
18. McGinnis LK, Luense LJ, Christenson LK. MicroRNA in ovarian biology and disease. Cold Spring Harb Perspect Med 2015;5:a022962.
crossref pmid pmc
19. Yang X, Zhou Y, Peng S, Wu L, Lin HY, Wang S, et al. Differentially expressed plasma microRNAs in premature ovarian failure patients and the potential regulatory function of mir-23a in granulosa cell apoptosis. Reproduction 2012;144:235-44.
crossref pmid
20. Ebrahimi SO, Reiisi S, Parchami Barjui S. Increased risk of polycystic ovary syndrome (PCOS) associated with CC genotype of miR-146a gene variation. Gynecol Endocrinol 2018;34:793-7.
crossref pmid
21. Jiang L, Huang J, Li L, Chen Y, Chen X, Zhao X, et al. MicroRNA-93 promotes ovarian granulosa cells proliferation through targeting CDKN1A in polycystic ovarian syndrome. J Clin Endocrinol Metab 2015;100:E729-38.
crossref pmid pmc
22. Naz MSG, Rahnemaei FA, Tehrani FR, Sayehmiri F, Ghasemi V, Banaei M, et al. Possible cognition changes in women with polycystic ovary syndrome: a narrative review. Obstet Gynecol Sci 2023;66:347-63.
crossref pmid pmc pdf
23. Goodman NF, Cobin RH, Futterweit W, Glueck JS, Legro RS, Carmina E, et al. American Association of Clinical Endocrinologists, American College of Endocrinology, and Androgen Excess and Pcos Society disease state clinical review: guide to the best practices in the evaluation and treatment of polycystic ovary syndrome--part 1. Endocr Pract 2015;21:1291-300.
crossref pmid
24. March WA, Moore VM, Willson KJ, Phillips DI, Norman RJ, Davies MJ. The prevalence of polycystic ovary syndrome in a community sample assessed under contrasting diagnostic criteria. Hum Reprod 2010;25:544-51.
crossref pmid
25. Goodarzi MO, Dumesic DA, Chazenbalk G, Azziz R. Polycystic ovary syndrome: etiology, pathogenesis and diagnosis. Nat Rev Endocrinol 2011;7:219-31.
crossref pmid pdf
26. Corbett S, Morin-Papunen L. The polycystic ovary syndrome and recent human evolution. Mol Cell Endocrinol 2013;373:39-50.
crossref pmid
27. de Wilde MA, Goverde AJ, Veltman-Verhulst SM, Eijkemans MJ, Franx A, Fauser BC, et al. Insulin action in women with polycystic ovary syndrome and its relation to gestational diabetes. Hum Reprod 2015;30:1447-53.
crossref pmid
28. Dumont A, Robin G, Catteau-Jonard S, Dewailly D. Role of anti-Müllerian hormone in pathophysiology, diagnosis and treatment of polycystic ovary syndrome: a review. Reprod Biol Endocrinol 2015;13:137.
crossref pmid pmc
29. Kosova G, Urbanek M. Genetics of the polycystic ovary syndrome. Mol Cell Endocrinol 2013;373:29-38.
crossref pmid pmc
30. Casarini L, Brigante G. The polycystic ovary syndrome evolutionary paradox: a genome-wide association studies-based, in silico, evolutionary explanation. J Clin Endocrinol Metab 2014;99:E2412-20.
crossref pmid
31. Dumesic DA, Oberfield SE, Stener-Victorin E, Marshall JC, Laven JS, Legro RS. Scientific statement on the diagnostic criteria, epidemiology, pathophysiology, and molecular genetics of polycystic ovary syndrome. Endocr Rev 2015;36:487-525.
crossref pmid pmc pdf
32. Patel S. Polycystic ovary syndrome (PCOS), an inflammatory, systemic, lifestyle endocrinopathy. J Steroid Biochem Mol Biol 2018;182:27-36.
crossref pmid
33. Imbar T, Eisenberg I. Regulatory role of microRNAs in ovarian function. Fertil Steril 2014;101:1524-30.
crossref pmid
34. Lomelí H, Ramos-Mejía V, Gertsenstein M, Lobe CG, Nagy A. Targeted insertion of Cre recombinase into the TNAP gene: excision in primordial germ cells. Genesis 2000;26:116-7.
crossref pmid
35. Baley J, Li J. MicroRNAs and ovarian function. J Ovarian Res 2012;5:8.
crossref pmid pmc
36. Ebrahimi SO, Reiisi S. Downregulation of miR-4443 and miR-5195-3p in ovarian cancer tissue contributes to metastasis and tumorigenesis. Arch Gynecol Obstet 2019;299:1453-8.
crossref pmid pdf
37. Cai S, Chen R, Li X, Cai Y, Ye Z, Li S, et al. Downregulation of microRNA-23a suppresses prostate cancer metastasis by targeting the PAK6-LIMK1 signaling pathway. Oncotarget 2015;6:3904-17.
crossref pmid pmc
38. Khan HA, Zhao Y, Wang L, Li Q, Du YA, Dan Y, et al. Identification of miRNAs during mouse postnatal ovarian development and superovulation. J Ovarian Res 2015;8:44.
crossref pmid pmc pdf
39. Sun XF, Li YP, Pan B, Wang YF, Li J, Shen W. Molecular regulation of miR-378 on the development of mouse follicle and the maturation of oocyte in vivo. Cell Cycle 2018;17:2230-42.
crossref pmid pmc
40. Donadeu FX, Schauer SN, Sontakke SD. Involvement of miRNAs in ovarian follicular and luteal development. J Endocrinol 2012;215:323-34.
crossref pmid
41. McBride D, Carré W, Sontakke SD, Hogg CO, Law A, Donadeu FX, et al. Identification of miRNAs associated with the follicular-luteal transition in the ruminant ovary. Reproduction 2012;144:221-33.
crossref pmid
42. Real FM, Sekido R, Lupiáñez DG, Lovell-Badge R, Jiménez R, Burgos M. A microRNA (mmu-miR-124) prevents Sox9 expression in developing mouse ovarian cells. Biol Reprod 2013;89:78.
pmid
43. Ro S, Song R, Park C, Zheng H, Sanders KM, Yan W. Cloning and expression profiling of small RNAs expressed in the mouse ovary. RNA 2007;13:2366-80.
crossref pmid pmc
44. Zhang J, Ji X, Zhou D, Li Y, Lin J, Liu J, et al. miR-143 is critical for the formation of primordial follicles in mice. Front Biosci (Landmark Ed) 2013;18:588-97.
crossref pmid
45. Zhang Q, Sun H, Jiang Y, Ding L, Wu S, Fang T, et al. MicroRNA-181a suppresses mouse granulosa cell proliferation by targeting activin receptor IIA. PLoS One 2013;8:e59667.
crossref pmid pmc
46. Moreno JM, Núñez MJ, Quiñonero A, Martínez S, de la Orden M, Simón C, et al. Follicular fluid and mural granulosa cells microRNA profiles vary in in vitro fertilization patients depending on their age and oocyte maturation stage. Fertil Steril 2015;104:1037-46e1.
crossref pmid
47. Shinoda G, De Soysa TY, Seligson MT, Yabuuchi A, Fujiwara Y, Huang PY, et al. Lin28a regulates germ cell pool size and fertility. Stem Cells 2013;31:1001-9.
crossref pmid pdf
48. Flemr M, Moravec M, Libova V, Sedlacek R, Svoboda P. Lin28a is dormant, functional, and dispensable during mouse oocyte-to-embryo transition. Biol Reprod 2014;90:131.
pmid
49. Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY. MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res 2007;67:8433-8.
crossref pmid pdf
50. Carletti MZ, Fiedler SD, Christenson LK. MicroRNA 21 blocks apoptosis in mouse periovulatory granulosa cells. Biol Reprod 2010;83:286-95.
pmid pmc
51. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 2008;18:997-1006.
crossref pmid pdf
52. Mori MA, Ludwig RG, Garcia-Martin R, Brandão BB, Kahn CR. Extracellular miRNAs: from biomarkers to mediators of physiology and disease. Cell Metab 2019;30:656-73.
crossref pmid pmc
53. Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res 2010;107:810-7.
crossref pmid
54. Long W, Zhao C, Ji C, Ding H, Cui Y, Guo X, et al. Characterization of serum microRNAs profile of PCOS and identification of novel non-invasive biomarkers. Cell Physiol Biochem 2014;33:1304-15.
crossref pmid pdf
55. Lanthier N, Leclercq IA. Adipose tissues as endocrine target organs. Best Pract Res Clin Gastroenterol 2014;28:545-58.
crossref pmid
56. Sang Q, Yao Z, Wang H, Feng R, Wang H, Zhao X, et al. Identification of microRNAs in human follicular fluid: characterization of microRNAs that govern steroidogenesis in vitro and are associated with polycystic ovary syndrome in vivo. J Clin Endocrinol Metab 2013;98:3068-79.
crossref pmid
57. Sirotkin AV, Lauková M, Ovcharenko D, Brenaut P, Mlyncek M. Identification of microRNAs controlling human ovarian cell proliferation and apoptosis. J Cell Physiol 2010;223:49-56.
crossref pmid
58. Chen YH, Heneidi S, Lee JM, Layman LC, Stepp DW, Gamboa GM, et al. miRNA-93 inhibits GLUT4 and is overexpressed in adipose tissue of polycystic ovary syndrome patients and women with insulin resistance. Diabetes 2013;62:2278-86.
crossref pmid pmc pdf
59. Murri M, Insenser M, Fernández-Durán E, SanMillán JL, Escobar-Morreale HF. Effects of polycystic ovary syndrome (PCOS), sex hormones, and obesity on circulating miRNA-21, miRNA-27b, miRNA-103, and miRNA-155 expression. J Clin Endocrinol Metab 2013;98:E1835-44.
crossref pmid
60. Mu J, Li Q. Anomalous expression of miR-103 in polycystic ovary syndrome influenced by hormonal, and metabolic variables. Exp Mol Pathol 2020;116:104482.
crossref pmid
61. Xiong W, Lin Y, Xu L, Tamadon A, Zou S, Tian F, et al. Circulatory microRNA 23a and microRNA 23b and polycystic ovary syndrome (PCOS): the effects of body mass index and sex hormones in an Eastern Han Chinese population. J Ovarian Res 2017;10:10.
crossref pmid pmc pdf
62. Sathyapalan T, David R, Gooderham NJ, Atkin SL. Increased expression of circulating miRNA-93 in women with polycystic ovary syndrome may represent a novel, non-invasive biomarker for diagnosis. Sci Rep 2015;5:16890.
crossref pmid pmc pdf
63. Rashad NM, Ateya MA, Saraya YS, Elnagar WM, Helal KF, Lashin ME, et al. Association of miRNA - 320 expression level and its target gene endothelin-1 with the susceptibility and clinical features of polycystic ovary syndrome. J Ovarian Res 2019;12:39.
crossref pmid pmc pdf
64. Deswal R, Dang AS. Dissecting the role of micro-RNAs as a diagnostic marker for polycystic ovary syndrome: a systematic review and meta-analysis. Fertil Steril 2020;113:661-9e2.
crossref pmid
65. Yuan WN, Tan L. MicroRNA-320 inhibits insulin resistance in patients with PCOS through regulating ERK1/2 signaling pathway. Biomed Res 2017;28:4946-9.

66. Flammer J, Konieczka K. Retinal venous pressure: the role of endothelin. EPMA J 2015;6:21.
crossref pmid pmc pdf
67. Jiang L, Li W, Wu M, Cao S. Ciculating miRNA-21 as a biomarker predicts polycystic ovary syndrome (PCOS) in patients. Clin Lab 2015;61:1009-15.
crossref pmid
68. Song DK, Sung YA, Lee H. The role of serum microRNA-6767-5p as a biomarker for the diagnosis of polycystic ovary syndrome. PLoS One 2016;11:e0163756.
crossref pmid pmc
69. Li Y, Xiang Y, Song Y, Wan L, Yu G, Tan L. Dysregulated miR-142, -33b and -423 in granulosa cells target TGFBR1 and SMAD7: a possible role in polycystic ovary syndrome. Mol Hum Reprod 2019;25:638-46.
crossref pmid pdf
70. Luense LJ, Veiga-Lopez A, Padmanabhan V, Christenson LK. Developmental programming: gestational testosterone treatment alters fetal ovarian gene expression. Endocrinology 2011;152:4974-83.
crossref pmid pmc
71. Zhang Y, Xu L. Preliminary study of Yulin mixture affecting the miR-320/SF-1/Cyp19a1 on mouse polycystic ovary syndrome model. Gynecol Endocrinol 2021;37:546-53.
crossref pmid
72. Xiang Y, Song Y, Li Y, Zhao D, Ma L, Tan L. miR-483 is down-regulated in polycystic ovarian syndrome and inhibits KGN cell proliferation via targeting insulin-like growth factor 1 (IGF1). Med Sci Monit 2016;22:3383-93.
crossref pmid pmc
73. Yang Y, Jiang H, Xiao L, Yang X. MicroRNA-33b-5p is overexpressed and inhibits GLUT4 by targeting HMGA2 in polycystic ovarian syndrome: an in vivo and in vitro study. Oncol Rep 2018;39:3073-85.
crossref pmid
74. Butler AE, Ramachandran V, Hayat S, Dargham SR, Cunningham TK, Benurwar M, et al. Expression of microRNA in follicular fluid in women with and without PCOS. Sci Rep 2019;9:16306.
crossref pmid pmc pdf
75. Sørensen AE, Wissing ML, Salö S, Englund AL, Dalgaard LT. MicroRNAs related to polycystic ovary syndrome (PCOS). Genes (Basel) 2014;5:684-708.
crossref pmid pmc
76. Scalici E, Traver S, Mullet T, Molinari N, Ferrières A, Brunet C, et al. Circulating microRNAs in follicular fluid, powerful tools to explore in vitro fertilization process. Sci Rep 2016;6:24976.
crossref pmid pmc pdf
77. Roth LW, McCallie B, Alvero R, Schoolcraft WB, Minjarez D, Katz-Jaffe MG. Altered microRNA and gene expression in the follicular fluid of women with polycystic ovary syndrome. J Assist Reprod Genet 2014;31:355-62.
crossref pmid pmc pdf
78. Naji M, Nekoonam S, Aleyasin A, Arefian E, Mahdian R, Azizi E, et al. Expression of miR-15a, miR-145, and miR-182 in granulosa-lutein cells, follicular fluid, and serum of women with polycystic ovary syndrome (PCOS). Arch Gynecol Obstet 2018;297:221-31.
crossref pmid pdf
79. Cai G, Ma X, Chen B, Huang Y, Liu S, Yang H, et al. MicroRNA-145 negatively regulates cell proliferation through targeting IRS1 in isolated ovarian granulosa cells from patients with polycystic ovary syndrome. Reprod Sci 2017;24:902-10.
crossref pmid pdf
80. Xu B, Zhang YW, Tong XH, Liu YS. Characterization of microRNA profile in human cumulus granulosa cells: Identification of microRNAs that regulate Notch signaling and are associated with PCOS. Mol Cell Endocrinol 2015;404:26-36.
crossref pmid
81. Lu J, Zhang C, Gu B, Zhang S, Geng J, Chen Y, et al. MicroRNA-182 inhibits rat ovarian granulosa cell apoptosis by targeting Smad7 in polycystic ovarian syndrome. Int J Clin Exp Pathol 2017;10:1380-7.

82. Xue Y, Lv J, Xu P, Gu L, Cao J, Xu L, et al. Identification of microRNAs and genes associated with hyperandrogenism in the follicular fluid of women with polycystic ovary syndrome. J Cell Biochem 2018;119:3913-21.
crossref pmid pdf
83. Wang M, Liu M, Sun J, Jia L, Ma S, Gao J, et al. MicroRNA-27a-3p affects estradiol and androgen imbalance by targeting Creb1 in the granulosa cells in mouse polycytic ovary syndrome model. Reprod Biol 2017;17:295-304.
crossref pmid
84. Zhang CL, Wang H, Yan CY, Gao XF, Ling XJ. Deregulation of RUNX2 by miR-320a deficiency impairs steroidogenesis in cumulus granulosa cells from polycystic ovary syndrome (PCOS) patients. Biochem Biophys Res Commun 2017;482:1469-76.
crossref pmid
85. Chen Z, Ou H, Wu H, Wu P, Mo Z. Role of microRNA in the pathogenesis of polycystic ovary syndrome. DNA Cell Biol 2019;38:754-62.
crossref pmid
86. Huang X, Liu C, Hao C, Tang Q, Liu R, Lin S, et al. Identification of altered microRNAs and mRNAs in the cumulus cells of PCOS patients: miRNA-509-3p promotes oestradiol secretion by targeting MAP3K8. Reproduction 2016;151:643-55.
crossref pmid
87. Tu J, Cheung AH, Chan CL, Chan WY. The role of microRNAs in ovarian granulosa cells in health and disease. Front Endocrinol (Lausanne) 2019;10:174.
crossref pmid pmc
88. Kang L, Yang C, Wu H, Chen Q, Huang L, Li X, et al. miR-26a-5p regulates TNRC6A expression and facilitates theca cell proliferation in chicken ovarian follicles. DNA Cell Biol 2017;36:922-9.
crossref pmid
89. Zielak-Steciwko AE, Browne JA, McGettigan PA, Gajewska M, Dzięcioł M, Szulc T, et al. Expression of microRNAs and their target genes and pathways associated with ovarian follicle development in cattle. Physiol Genomics 2014;46:735-45.
crossref pmid
90. Donadeu FX, Mohammed BT, Ioannidis J. A miRNA target network putatively involved in follicular atresia. Domest Anim Endocrinol 2017;58:76-83.
crossref pmid pmc
91. Yang R, Chen J, Wang L, Deng A. LncRNA BANCR participates in polycystic ovary syndrome by promoting cell apoptosis. Mol Med Rep 2019;19:1581-6.
crossref pmid pmc
92. Zhang BB, Li XN, Li MX, Sun YY, Shi YX, Ma TH. miR-140-3p promotes follicle granulosa cell proliferation and steroid hormone synthesis via targeting AMH in chickens. Theriogenology 2023;202:84-92.
crossref pmid
93. Hossain MM, Cao M, Wang Q, Kim JY, Schellander K, Tesfaye D, et al. Altered expression of miRNAs in a dihydrotestosterone-induced rat PCOS model. J Ovarian Res 2013;6:36.
crossref pmid pmc pdf
94. Crespo RP, Bachega TASS, Mendonça BB, Gomes LG. An update of genetic basis of PCOS pathogenesis. Arch Endocrinol Metab 2018;62:352-61.
crossref pmid pmc
95. De Nardo Maffazioli G, Baracat EC, Soares JM, Carvalho KC, Maciel GAR. Evaluation of circulating microRNA profiles in Brazilian women with polycystic ovary syndrome: a preliminary study. PLoS One 2022;17:e0275031.
crossref pmid pmc
96. Mathivanan S, Simpson RJ. ExoCarta: a compendium of exosomal proteins and RNA. Proteomics 2009;9:4997-5000.
crossref pmid pdf
97. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. The microRNA spectrum in 12 body fluids. Clin Chem 2010;56:1733-41.
crossref pmid pmc pdf
98. Lässer C, Alikhani VS, Ekström K, Eldh M, Paredes PT, Bossios A, et al. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med 2011;9:9.
pmid pmc
99. Andre F, Schartz NE, Movassagh M, Flament C, Pautier P, Morice P, et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet 2002;360:295-305.
crossref pmid
100. Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Cabo F, Pérez-Hernández D, Vázquez J, Martin-Cofreces N, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun 2013;4:2980.
crossref pmid pmc pdf
101. Shurtleff MJ, Temoche-Diaz MM, Karfilis KV, Ri S, Schekman R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. Elife 2016;5:e19276.
crossref pmid pmc pdf
102. McKenzie AJ, Hoshino D, Hong NH, Cha DJ, Franklin JL, Coffey RJ, et al. KRAS-MEK signaling controls ago2 sorting into exosomes. Cell Rep 2016;15:978-87.
crossref pmid pmc
103. Che X, Jian F, Chen C, Liu C, Liu G, Feng W. PCOS serum-derived exosomal miR-27a-5p stimulates endometrial cancer cells migration and invasion. J Mol Endocrinol 2020;64:1-12.
crossref pmid
104. Hong Y, Wu J, Yu S, Hui M, Lin S. Serum-derived exosomal microRNAs in lipid metabolism in polycystic ovary syndrome. Reprod Sci 2022;29:2625-35.
crossref pmid pdf
105. Zhang F, Li SP, Zhang T, Yu B, Zhang J, Ding HG, et al. High throughput microRNAs sequencing profile of serum exosomes in women with and without polycystic ovarian syndrome. PeerJ 2021;9:e10998.
crossref pmid pmc pdf
106. Jiang X, Li J, Zhang B, Hu J, Ma J, Cui L, et al. Differential expression profile of plasma exosomal microRNAs in women with polycystic ovary syndrome. Fertil Steril 2021;115:782-92.
crossref pmid
107. Sang Q, Yao Z, Wang H, Feng R, Wang H, Zhao X, et al. Identification of microRNAs in human follicular fluid: characterization of microRNAs that govern steroidogenesis in vitro and are associated with polycystic ovary syndrome in vivo. J Clin Endocrinol Metab 2013;98:3068-79.
crossref pmid
108. Sohel MMH, Hoelker M, Schellander K, Tesfaye D. The extent of the abundance of exosomal and non-exosomal extracellular miRNAs in the bovine follicular fluid. Reprod Domest Anim 2022;57:1208-17.
crossref pmid pdf
109. Li H, Huang X, Chang X, Yao J, He Q, Shen Z, et al. S100-A9 protein in exosomes derived from follicular fluid promotes inflammation via activation of NF-κB pathway in polycystic ovary syndrome. J Cell Mol Med 2020;24:114-25.
crossref pmid pmc pdf
110. Santonocito M, Vento M, Guglielmino MR, Battaglia R, Wahlgren J, Ragusa M, et al. Molecular characterization of exosomes and their microRNA cargo in human follicular fluid: bioinformatic analysis reveals that exosomal microRNAs control pathways involved in follicular maturation. Fertil Steril 2014;102:1751-61e1.
crossref pmid
111. Hu J, Tang T, Zeng Z, Wu J, Tan X, Yan J. The expression of small RNAs in exosomes of follicular fluid altered in human polycystic ovarian syndrome. PeerJ 2020;8:e8640.
crossref pmid pmc pdf
112. Yang Q, Liu L, Huang H. Extraction and identification of exosomes in follicular fluid from patients with polycystic ovary syndrome and isolation and detection of miRNAs in exosomes. J Shanghai Jiaotong Univ (Med Sci) 2017;38:1085-9.

113. Yuan D, Luo J, Sun Y, Hao L, Zheng J, Yang Z. PCOS follicular fluid derived exosomal miR-424-5p induces granulosa cells senescence by targeting CDCA4 expression. Cell Signal 2021;85:110030.
crossref pmid
114. Zhao Y, Pan S, Li Y, Wu X. Exosomal miR-143-3p derived from follicular fluid promotes granulosa cell apoptosis by targeting BMPR1A in polycystic ovary syndrome. Sci Rep 2022;12:4359.
crossref pmid pmc pdf
115. Zhou Z, Tu Z, Zhang J, Tan C, Shen X, Wan B, et al. Follicular fluid-derived exosomal microRNA-18b-5p regulates PTEN-mediated PI3K/Akt/mTOR signaling pathway to inhibit polycystic ovary syndrome development. Mol Neurobiol 2022;59:2520-31.
crossref pmid pdf
116. Hanna J, Hossain GS, Kocerha J. The potential for microRNA therapeutics and clinical research. Front Genet 2019;10:478.
crossref pmid pmc
117. Johnson AL. Intracellular mechanisms regulating cell survival in ovarian follicles. Anim Reprod Sci 2003;78:185-201.
crossref pmid
118. Yang MY, Rajamahendran R. Morphological and biochemical identification of apoptosis in small, medium, and large bovine follicles and the effects of follicle-stimulating hormone and insulin-like growth factor-I on spontaneous apoptosis in cultured bovine granulosa cells. Biol Reprod 2000;62:1209-17.
pmid
119. Das M, Djahanbakhch O, Hacihanefioglu B, Saridogan E, Ikram M, Ghali L, et al. Granulosa cell survival and proliferation are altered in polycystic ovary syndrome. J Clin Endocrinol Metab 2008;93:881-7.
crossref pmid pmc pdf
120. Weroha SJ, Haluska P. The insulin-like growth factor system in cancer. Endocrinol Metab Clin North Am 2012;41:335-50. -vi.
crossref pmid pmc
121. Geng Y, Sui C, Xun Y, Lai Q, Jin L. MiRNA-99a can regulate proliferation and apoptosis of human granulosa cells via targeting IGF-1R in polycystic ovary syndrome. J Assist Reprod Genet 2019;36:211-21.
crossref pmid pmc pdf
122. Wang T, Liu Y, Lv M, Xing Q, Zhang Z, He X, et al. miR-323-3p regulates the steroidogenesis and cell apoptosis in polycystic ovary syndrome (PCOS) by targeting IGF-1. Gene 2019;683:87-100.
crossref pmid
123. Dekel N, Beers WH. Development of the rat oocyte in vitro: inhibition and induction of maturation in the presence or absence of the cumulus oophorus. Dev Biol 1980;75:247-54.
crossref pmid
124. Salehi E, Aflatoonian R, Moeini A, Yamini N, Asadi E, Khosravizadeh Z, et al. Apoptotic biomarkers in cumulus cells in relation to embryo quality in polycystic ovary syndrome. Arch Gynecol Obstet 2017;296:1219-27.
crossref pmid pdf
125. Bakhshalizadeh S, Amidi F, Alleyassin A, Soleimani M, Shirazi R, Shabani Nashtaei M. Modulation of steroidogenesis by vitamin D3 in granulosa cells of the mouse model of polycystic ovarian syndrome. Syst Biol Reprod Med 2017;63:150-61.
crossref pmid
126. Yang HS, Cho MH, Zakowicz H, Hegamyer G, Sonenberg N, Colburn NH. A novel function of the MA-3 domains in transformation and translation suppressor Pdcd4 is essential for its binding to eukaryotic translation initiation factor 4A. Mol Cell Biol 2004;24:3894-906.
crossref pmid pmc pdf
127. Ding L, Gao F, Zhang M, Yan W, Tang R, Zhang C, et al. Higher PDCD4 expression is associated with obesity, insulin resistance, lipid metabolism disorders, and granulosa cell apoptosis in polycystic ovary syndrome. Fertil Steril 2016;105:1330-7e3.
crossref pmid
128. Zhao Y, Tao M, Wei M, Du S, Wang H, Wang X. Mesenchymal stem cells derived exosomal miR-323-3p promotes proliferation and inhibits apoptosis of cumulus cells in polycystic ovary syndrome (PCOS). Artif Cells Nanomed Biotechnol 2019;47:3804-13.
crossref pmid
129. Wei Y, Lu S, Hu Y, Guo L, Wu X, Liu X, et al. MicroRNA-135a regulates VEGFC expression and promotes luteinized granulosa cell apoptosis in polycystic ovary syndrome. Reprod Sci 2020;27:1436-42.
crossref pmid pdf
130. Bago R, Sommer E, Castel P, Crafter C, Bailey FP, Shpiro N, et al. The hVps34-SGK3 pathway alleviates sustained PI3K/Akt inhibition by stimulating mTORC1 and tumour growth. EMBO J 2016;35:2263.
pmid pmc
131. Wang W, Dong J, Wang M, Yao S, Tian X, Cui X, et al. miR-148a-3p suppresses epithelial ovarian cancer progression primarily by targeting c-Met. Oncol Lett 2018;15:6131-6.
crossref pmid pmc
132. La Marca A, Broekmans FJ, Volpe A, Fauser BC, Macklon NS; ESHRE Special Interest Group for Reproductive Endocrinology--AMH Round Table. Anti-Mullerian hormone (AMH): what do we still need to know? Hum Reprod 2009;24:2264-75.
crossref pmid
133. Díaz M, Bassols J, López-Bermejo A, de Zegher F, Ibáñez L. Low circulating levels of miR-451a in girls with polycystic ovary syndrome: different effects of randomized treatments. J Clin Endocrinol Metab 2020;105:dgz204.
pmid
134. Chen WH, Huang QY, Wang ZY, Zhuang XX, Lin S, Shi QY. Therapeutic potential of exosomes/miRNAs in polycystic ovary syndrome induced by the alteration of circadian rhythms. Front Endocrinol (Lausanne) 2022;13:918805.
crossref pmid pmc
135. Anna G, Kannan NN. Post-transcriptional modulators and mediators of the circadian clock. Chronobiol Int 2021;38:1244-61.
crossref pmid pmc
136. Torres M, Becquet D, Franc JL, François-Bellan AM. Circadian processes in the RNA life cycle. Wiley Interdiscip Rev RNA 2018;9:e1467.
crossref pmid pdf
137. Tao SC, Guo SC. Extracellular vesicles: potential participants in circadian rhythm synchronization. Int J Biol Sci 2018;14:1610-20.
crossref pmid pmc


ABOUT
ARTICLE & TOPICS
Article category

Browse all articles >

Topics

Browse all articles >

BROWSE ARTICLES
POLICY
FOR CONTRIBUTORS
Editorial Office
4th Floor, 36 Gangnam-daero 132-gil, Gangnam-gu, Seoul 06044, Korea.
Tel: +82-2-2266-7238    Fax: +82-2-3445-2440    E-mail: journal@ogscience.org                

Copyright © 2024 by Korean Society of Obstetrics and Gynecology.

Developed in M2PI

Close layer
prev next