Abstract
Purpose
This review aims to summarize the key findings of several miRNAs and their roles in polycystic ovary syndrome with insulin resistance, characterize the disease pathogenesis, and establish a new theoretical basis for diagnosing, treating, and preventing polycystic ovary syndrome.
Methods
Relevant scientific literature was covered from 1992 to 2020 by searching the PubMed database with search terms: insulin/insulin resistance, polycystic ovary syndrome, microRNAs, and metabolic diseases. References of relevant studies were cross-checked.
Results
The related miRNAs (including differentially expressed miRNAs) and their roles in pathogenesis, and possible therapeutic targets and pathways, are discussed, highlighting controversies and offering thoughts for future directions.
Conclusion
We found abundant evidence on the role of differentially expressed miRNAs with its related phenotypes in PCOS. Considering the essential role of insulin resistance in the pathogenesis of PCOS, the alterations of associated miRNAs need more research attention. We speculate that race/ethnicity or PCOS phenotype and differences in methodological differences might lead to inconsistencies in research findings; thus, several miRNA profiles need to be investigated further to qualify for the potential therapeutic targets for PCOS-IR.
Keywords: miRNAs, PCOS, Insulin resistance, Adipose, Oocyte quality
Introduction
Polycystic ovary syndrome (PCOS) is a reproductive endocrine disorder, which affects 12% of women at childbearing age [1]. The main characteristics include polycystic ovaries, hyperandrogenism, chronic anovulation, and insulin resistance (IR), despite their highly heterogeneous features [2]. PCOS is associated with various diseases, including anovulatory infertility [3] and metabolic disorders such as metabolic syndrome, type 2 diabetes, and cardiovascular disease in women [4, 5]. The etiology of PCOS may include genetic [6], environmental, and epigenetic factors [7]. IR refers to the decreased sensitivity of peripheral tissue to insulin and thus impairing glucose uptake and production. The insufficient glucose consumption results in blood glucose level upregulation and stimulates the pancreatic β-cells of the islet to secret excessive insulin to lower blood glucose [8], which leads to hyperinsulinism. More than 60% of PCOS patients have varying degrees of IR [9], and their insulin intake efficiency is 20%–40% lower than that in healthy people [10]. IR and compensatory hyperinsulinism drive multiple phenotypic characteristics in PCOS [2], mainly because insulin facilitates androgen secretion predominately by increasing the synthesis and release of ovarian androgens [2, 11] and modulating luteinizing hormone secretion from the pituitary [12]. Thus, hyperinsulinism contributes to clinical manifestations, including ovulatory disturbances [13], menstrual irregularity, anovulatory infertility, and immature follicles increase [14] in PCOS. Clinically, IR is mostly detected and assessed by the homeostasis model assessment of insulin resistance (HOMA-IR) in vivo, and the precision is equivalent to the glucose clamp technique. However, there is great variability in the threshold HOMA-IR levels to define IR [15]. Research showed that only 49.6% of PCOS patients were diagnosed as IR; however, 22.6% and 15.8% of patients were diagnosed as IR when the blood glucose cutoff value was set to 3.46 and 3.8 using HOMA-IR [16]. Another study found that the HOMA-IR index increased with age, stabilized between 13 and 15 years old, and then began to decline year by year among Caucasians [17]. Therefore, the diagnosis of PCOS-IR by HOMA-IR is affected by demographics and setting different cutoff values may generate different results. We recognize the potential validity issue due to the judgment criteria. However, for this review, studies have set similar HOMA-IR threshold index values (approximately between 2.2 and 2.75). Thus, we assume that the heterogeneity of the population does not affect the validity of this review.
miRNAs are small non-coding, single-stranded RNAs with 19–25 nucleotides in length. As post-transcriptional regulators of gene expression, miRNA is involved in suppressing mRNA expression and inhibiting protein translation by binding to the 3′ UTR region of the target mRNAs [18]. Also, miRNAs may influence the expression and alter their downstream target gene [19] and affect cell growth, differentiation, proliferation and apoptosis, inflammation, stress response [20], and malignant tumor metastasis. Many studies have shown that alteration of miRNAs may contribute to the onset of cervical cancer [21], endometriosis [22], poor ovarian response, and cardiovascular diseases [23, 24], and thus can serve as potential diagnostic and monitoring biomarkers in various diseases [25], including diabetes [26] and PCOS [27]. Among PCOS patients, several miRNAs are differentially expressed in serum [28], adipose tissue (AT) [29], follicular fluid (FF), granulosa cells (GCs) [30, 31], and cumulus cells (CCs). Certain miRNAs may be new biomarkers for abnormal metabolism, compromised oocyte quality, and low endometrial receptivity in PCOS [32].
The etiologies of miRNAs alteration in PCOS are not well defined yet. Most research has focused on the dysregulation of miRNAs in PCOS and healthy people [33, 34] and has shown that miRNAs might play an important role in the occurrence and development of PCOS-IR and PCOS non-insulin resistant (PCOS-NIR) [35, 36]. Some studies have found the key differences between PCOS-IR and PCOS-NIR in various metabolic indexes, including significant differences in molecules involved in insulin signaling, IGF system, lipid metabolism, and steroidogenic signaling such as PPAR-γ, CYP19A1, and IRS [37]. In addition, assisted reproductive technology (ART) outcomes were compared between PCOS-IR and PCOS-NIR cohort. Studies found that PCOS-IR patients have lower pregnancy rate, number of oocytes retrieved [38], ovulation rate, pregnancy rate, and worse outcome in ovulation induction therapy. Besides, PCOS-IR patients are associated with increased pregnancy complications, including gestational diabetes and pregnancy hypertension, and adverse pregnancy outcomes including miscarriage and premature births [39]. The aim of this paper is to review relevant literature to find the crucial miRNAs in the pathogenesis of PCOS-IR and explore the possible mechanisms in the insulin signaling pathways (Fig. 1). This review highlights the role of miRNAs in PCOS and might provide novel insights on biomarkers and treatments for PCOS-IR.
Fig. 1.
The roles of miRNAs in the insulin signaling pathways and insulin resistance in PCOS
Alterations of miRNAs expression of PCOS-IR in different tissues
miRNA is highly complex and is associated with insulin resistance in PCOS patients. Therefore, we looked into the differences in the expression of miRNAs between PCOS-IR and PCOS-NIR, especially in different tissues. With the development of gene sequencing technology, many researchers have carried out miRNA sequencing studies on PCOS with various samples such as follicular fluid, plasma, granulosa cells, and cumulus cells. Since the miRNAs produced at a peripheral tissue are not all be released into the blood, determining the tissue difference of miRNA expression level will indicate the tissue-specific accumulation and reflect the reliability sample collection to get accurate results.
Differential expression of miRNAs in serum
miRNAs are abundant and stable in human serum [40]. Since serum miRNAs are easy to be detected, potentially disease-specific, and have little individual expression differences, they attracted most research attention and became ideal biomarkers [41]. A study found that miR-222, miR-146a, and miR-30c were differentially expressed in the serum of five women with PCOS and five healthy people [28]. Among them, miR-222 was positively associated with serum insulin. Of note, miR-222 was significantly increased in type 2 diabetes and may be related to insulin sensitivity [42]. miR-146a was negatively associated with serum testosterone, and it was also reported to inhibit the release of progesterone, androgens, and estrogens. It is speculated that miR-146a may act as a physiological inhibitor of general secretory activity [43]. In addition, bioinformatics analyses indicate that genes targeted by miR-222, miR-146a, and miR-30 are involved in the cell cycle, apoptosis, and endocrine pathways that participate in the pathogenesis of PCOS via insulin-related signaling pathways, including Wnt signaling pathway and Jak-STAT signaling pathway [28]. A study revealed that the serum levels of miR-24 and miR-29a were lowered in PCOS subjects and could be of diagnostic value [44]. Another study [45] found that miR-24 level in the plasma of type 2 diabetic patients was significantly lower than that in healthy controls and the insulin promoter activity was found to be downregulated by miR-24, thereby resulting in the reduction in insulin mRNA levels. Androgen receptor (AR), StAR-related lipid transfer protein3 (STARD3), and insulin receptor substrate-1 (IRS-1) are reported as targets of miR-29a. Among these, AR and STARD3 are involved in ovarian steroidogenesis, while IRS-1 is involved in insulin signaling.
Jiang et al. found 748 miRNAs expression profiles of three pooled samples (10 PCOS-NGT samples, 10 PCOS-IGT samples, and 10 controls) from the serum of 175 women, among which three miRNAs (miR-122, miR-193b, miR-194) were differentially expressed between PCOS-IGT (impaired glucose tolerance) and PCOS-NGT (normal glucose tolerance) samples [46]. Wang et al. demonstrated that miR-122 was a potential biomarker of obesity and IR [47]. miR-122 and miR-193b expression levels were significantly higher in diabetic cats compared with those of healthy cats [46]. Overexpressed miR-194 in the muscle and liver elevated fasting glucose levels in rats by targeting the IGF-1 receptor [31], suggesting its functions in the regulation of glucose metabolism. Another study [48] detected the expression of miR-204 and analyzed the diagnostic efficacy in the serum of between PCOS-IR patients and PCOS-NIR patients and found that both in vitro and in vivo experiments supported the capabilities of miR-204 and HMGB1 in proliferation and apoptosis of PCOS-IR GCs. In addition, it was revealed that overexpression of miR-204 and downregulation of HMGB1 inhibited TLR4/NF-kB pathway activation, decrease IR and T, and increase ovarian coefficient. It has been suggested previously that overexpression of miR-204 restrains the JAK2/STAT3 pathway [49].
Differential expression of miRNAs in follicular fluid
Follicular fluid provides an appropriate intrafollicular environment for oocytes maturation and development, which comprises the secretory and metabolic activity product of the oocyte, GCs, theca cells (TCs), and the transfer of blood plasma components. Therefore, the analysis of FF ingredients may also reflect metabolic and functional changes in the oocyte and GCs [50]. Oocytes are deemed as high quality when they undergo meiosis and achieve fertilization, followed by embryogenesis and development. Various pathological conditions that disrupt the intrafollicular environment may affect the quality of oocytes, the development of the early embryo, and the outcome of the pregnancy [50, 51]. A study has shown that there are abnormalities in the components of the FF in PCOS patients. Furthermore, alterations in blood circulation biochemical components may suggest changes in the composition of FF.
Roth et al. found that 5 miRNAs (32, 34c, 135a, 18b, and 9) showed significantly increased expression in the PCOS group, and pathway analysis revealed 3 potential genes (IL-8, SYT1, and IRS-2) involved in insulin regulation and inflammation [52]. IRS-2 mediates the effects of insulin and IGF-1 and is critical for peripheral carbohydrate metabolism and beta-cell function. Mice lacking IRS-2 developed estrous cycle disorders, anovulation, infertility, and IR, similar to women with PCOS [53]. SYT1 is a gene encoding synaptogamin1 and a calcium-binding synaptic vesicle protein [54], and it plays a critical role in calcium-triggered exocytosis throughout the body and is required for exocytosis of insulin in the pancreatic beta-cells. Compared to the healthy group, miR-1290 was one of the highest expressed miRNAs in FF of PCOS [55] and also highly expressed in gestational diabetes [56]. Sorensen et al. also found that the increased miR-1290 is associated with either aberrant glucose homeostasis or dyslipidemia [57], indicating that high levels of miR-1290 may impact glucose and insulin metabolism and act on PCOS pathophysiology. In other studies, the expression of miR-320 is different in the FF with PCOS women [55, 58], indicating that miR-320 might serve as a potential target to improve insulin sensitivity and IR. FF is easily obtained from in vitro fertilization and oocyte harvesting procedures, which becomes an ideal setting to investigate the oocyte quality, maturation, and fertility outcomes; thus, all the above miRNAs have the potential to be developed as new biomarkers for PCOS-IR.
Differential expression of miRNAs in other tissues
IR presents ubiquitously throughout the body and is associated with not only PCOS but also other health issues, including obesity and cardiovascular problems. Several studies revealed the differential expression of miRNAs in other tissues related to IR. In adipocytes of PCOS, the upregulation of miRNA-93 and miRNA-223 downregulated GLUT4 gene expression, which showed that miRNA-93 and miRNA-223 might be associated with IR in PCOS [59]. Subsequently, miRNA-93 in AT correlated with PCOS-IR though its host gene MCM7 was discordantly downregulated [29]. In plasma of PCOS, miRNA-93 and miRNA-223 expressions were also increased, but miRNA levels were independent of insulin and IR [60]. In ovarian theca tissues, the downregulation of miR-92a and miR-92b might lead to augmentation of signal transduction in the insulin pathway between PCOS patients and controls [36]. In the theca cells, Lin et al. reported that the expression of miR-92a and miR-92b was significantly downregulated with PCOS. In another study, GATA6 and IRS-2 exhibited significantly higher PCOS TCs, which is in agreement with Lin et al.’s findings. Also, GATA6 was predicted as a target gene of miR-92a and IRS-2 was regulated by both miR-92a and miR-92b [61]. In ovaries from PCOS-IR rats [62], miR-30 was significantly upregulated, which is consistent with previous studies [28, 35]. However, miR-146 was significantly upregulated in serum from PCOS patients compared with that of healthy women, which is inconsistent with the results in rats [63]. This difference may be explained by the differences of species, natural heterogeneity of PCOS, and/or the different sequencing method employed.
Common miRNAs in different tissues in PCOS-IR
Although the miRNAs in different tissues are diverse in PCOS-IR, there are some commonly expressed miRNAs in different tissues. miR-320a is an inhibitor related to various kinds of tumors, as its abnormal expression is mostly associated with malignant tumors [64]. In the female reproductive system, the expression level of miR-320a is closely related to the development of oocytes, and maintaining a normal expression level is necessary for oocyte development. The research reported that mature oocytes were associated with a higher level of miR-320a [65]. However, miR-320a was expressed at significantly lower levels in serum and FF of PCOS-IR patients than in healthy controls [55]. Similarly, the serum level of miRNA-320 was also reduced in the diabetic population [66] and adipocytes [67], and studies have speculated that miR-320a may reduce insulin sensitivity through the insulin PI3K signaling pathway [68, 69]; therefore, miRNA-320 plays a key role in regulating IR. RAB5B, identified as a PCOS candidate gene in a genome-wide association study, is predicted to be the target gene of miRNA-320. We may postulate that lower miRNA-320 expression levels in the FF of PCOS patients may affect RAB5B gene expression. miR-320 can also reduce the expression of IRS and inhibit the phosphorylation of extracellular-regulated protein kinases (ERK1/2) pathway which may lead to IR [70]. However, the expression of miR-320 was unregulated in AT of PCOS-IR [71]. Some studies have found different miRNA expression results. The research reported the upregulation of miRNA-320 in FF, potentially explaining the hyperandrogenemia found in women with PCOS through downregulation of E2F1/SF-1 proteins, which in turn cause the inhibition of estradiol releasing into FF [58]. Another study showed that there is no differential expression of either miR-132 or 320 in FF of PCOS patients in the USA [52]. Of note, serum miRNA-132 levels are significantly decreased in gestational diabetes mellitus suggesting that it may regulate insulin secretion [72]. This may be related to race/ethnicity or PCOS phenotype differences as Sang et al.’s study [55] population was all Asian, and they used genome-wide analysis in addition to microarray analysis to identify miRNAs, so methodological differences can also explain the differences.
miR-145 is elevated in FF, while their equivalents in GCs showed diminished expression in recent research [73]. In GCs of mice, miR-145 was found to be in the suppression of proliferation [74]. The decreased level of miR-145 in GCs of PCOS patients and its contribution to negative regulation of proliferation via targeting IRS1 have been studied, and miR-145 overexpression was found to suppress cell survival rate and DNA synthesis levels in human GCs [75]. In the ovarian cortex, miR-483 was downregulated in PCOS patients and dual-luciferase reporter assay showed IGF1 was a direct target for miR-483. Insulin treatment in KGN cells inhibits miR-483, promotes IGF1, and induces cell proliferation. miR-483-3p was confirmed to inhibit KGN cell proliferation and arrest cell cycle, possibly via suppressing IGF1, suggesting the potential to use miR-483 as a biomarker and therapeutic alternative for diagnosing and treating PCOS [76]. In CCs, qRT-PCR (quantitative real-time PCR) validated that miR-483–5p was downregulated in patients with PCOS [77]. miR-483–5p is a conserved sequence encoded in the second intron of the IGF2 gene [78]. IGF2, as a miR-483–5p co-expressed gene [79], was significantly downregulated in CCs with PCOS, which may be also one of the pathogenic factors for PCOS. In another study, miR-483-5p was identified to regulate the Notch and MAPK signaling pathway. miR-483-5p expression was increased in PCOS CCs, whereas the expression of MAPK3 mRNA and protein was decreased as shown in deep sequencing and qRT-PCR. Moreover, miR-483-5p regulated MAPK3 expression by directly binding 3′ UTR of mRNA based on the bioinformatics prediction and experimental validation [30]. A study has found that miR-483-5p can promote the proliferation of GCs in PCOS and play an important role in reducing IR [77].
miRNAs and glucose metabolism in PCOS-IR
Many differentially expressed miRNAs in PCOS women are involved in pathological insulin signaling processes, the disorder secretion of inflammatory factors, and hormones [80]. Hyperinsulinism may further contribute to the PCOS pathology. Nowadays, the relationships among miRNAs, PCOS, and IR have been assessed at transcript, genomic, and metabolic pathway levels. miRNAs participate in the insulin signaling process through activation of the insulin receptor which increases phosphorylation of intracellular substrates (IRS), principally related IRS proteins (IRS1, IRS2). For example, miR-145 directly inhibited IRS1 expression by binding to its 3′ UTR and the IRS1 overexpression abrogates the suppressive effect of miR-145 mimics. The overexpression of miR-145 inhibited MAKP/ERK signaling pathways in GCs. A high concentration of insulin can reduce miR-145 expression, upregulate IRS1 protein expression, and promote cell proliferation [75]. These studies indicate that the expression of miR-145 may not only inhibit GC proliferation but also improve IR in PCOS patients. In addition, miR-370 is a scaffold protein involved in the insulin pathway modulating IRS1 expression [81] and the downregulation of miR-370 may target IRS1 and disturb insulin metabolism of GCs and affect oocyte quality [82, 83]. Table 1 shows the literature that summarized the expression and function of miRNAs among PCOS-IR patients.
Table 1.
Related microRNAs observed in polycystic ovary syndrome (PCOS) with insulin resistance
miRNA | Species | Samples | Criterions of IR | Expression | Targets and pathways | Proposed functions | Ref. |
---|---|---|---|---|---|---|---|
miR-204 | Human | Serum | HOMA-IR > 2.57 | Decreased | HMGB1; TLR4/NF-KB pathway | Reduces IR, boosts proliferation, restrains apoptosis | [48] |
miR-612 | Human | Cumulus cells | HOMA-IR > 2.2 | Decreased | Rap1b; MAPK pathway | Involved in pathophysiology | [84] |
miR-93 | Human | Adipose tissue | HOMA-IR ≥ 2.5 | Increased | Glut4; IRS1/PI3K/AKT pathway | Related to AT metabolism | [59] |
Human | Serum | - | Increased | — | Can be a biomarker potentially | [60] | |
miR-223 | Human | Adipose tissue | HOMA-IR ≥ 2.5 | Increased | Glut4 | Inhibits Glut4 expression and induces IR | [85] |
miR-33b-5p | Rat | Ovarian tissue | HOMA-IR ≥ 2.5 | Increased | Hmga2; PI3K/Akt | Inhibits GLUT4 production by targeting HMGA2 | [86] |
miR-92a | Human | Ovarian tissues | HOMA-IR > 2.14 | Decreased | GATA6, IRS-2 | Related to the pathogenesis of PCOS | [36] |
miR25/93/106b | Human | Adipose tissue | HOMA-IR ≥ 2.5 | Increased | MCM7 | May not be regulated by target gene MCM7 | [29] |
miR-485-3p | Human | Skeletal muscle | Hyperinsulinemic-euglycemic clamp | Increased | PGC-1α | Inhibits PGC-1α and impart IR in PCOS | [87, 88] |
miR-93 | Human | Ovary cortex | HOMA-IR ≥ 2.14 | Increased | CDKN1A | Promotes cell proliferation | [89] |
miR-451a | Human | Serum | — | Decreased | PI3K/AKT pathway | A novel candidate biomarker for PCOS diagnosis | [90] |
miR-483-5p | Human | Cumulus cells | — | Decreased | IGF2 | Reduces IR in PCOS | [77] |
miR-486-5p | Human | Cumulus cells | — | Decreased | PTEN, DOCK3; PI3K/Akt pathway | Promotes the proliferation of CCs | [77] |
miR-323-3p | Human | Cumulus cells | — | Decreased | IGF1; Wnt and TGF-β pathway | Inhibits steroidogenesis and CCs apoptosis | [91] |
Human | Follicular fluid | Not detected | [55] | ||||
miR-222 | Human | Serum | — | Increased | — | Correlated to serum insulin in PCOS | [28] |
miR-378a-5p | Human | Serum | — | Increased | PGC-1β, PI3K pathway | Acts between PCOS-HA and IR | [57, 92] |
miR-15b | Human | Adipose tissue | — | Decreased | Dicer | Be important in adipose differentiation | [93] |
miR-320 | Human | Follicular fluid | — | Decreased | RAB5B | Regulates IR in PCOS women | [55] |
miR-320a | Human | Ovarian tissues | — | Increased | PCGF1 | Reduces KGN cell proliferation and apoptosis | [94] |
Human | Follicular fluid | Increased | E2F1/SF-1 | Causes inhibition of estradiol release into FF | [58] | ||
miR-193b | Human | Serum | — | Increased | — | Correlated with HOMA-IR | [46] |
miR-122 | Human | Serum | — | Increased | — | A new biomarker of obesity and IR possibly | [46, 47] |
miR-194 | Human | Serum | — | Increased | IGF-1 | Regulates glucose metabolism | [46, 95] |
miR-145 | Human | Granulosa cells | — | Decreased | IRS-1; MAPK/ERK pathway | Improve IR in PCOS | [75] |
Human | Follicular fluid | Increased | — | Involved in pathogenesis of PCOS | [31] | ||
miR-27a-3p | Human | Granulosa cells | — | Decreased | STAT1, STAT3; SMAD5 | Decreases proliferation and promotes apoptosis | [96] |
miR-140 | Human | Granulosa cells | — | Increased | RAP2A; RAP2A/AKT pathway | Increases proliferation and reduces apoptosis | [97] |
miR-483 | Human | Ovary cortex | — | Decreased | IGF1 | Inhibits KGN cell proliferation | [76] |
miR-9119 | Rat | Granulosa cells | — | Decreased | p65, DICER; NF-κB/p65 pathway | Inhibits cell proliferation and promotes apoptosis | [98] |
Glucose enters the cell membrane mainly through glucose transporter 4 (GLUT4) [99], which is an insulin-stimulated glucose transporter and one of the most important candidate genes for IR. The expression level of GLUT4 is positively correlated with glucose translocation [100] tolerance as well as insulin sensitivity [101] and its lower expression will impair the glucose metabolism process [102]. The expression of GLUT4 is detected in AT from PCOS-IR, PCOS, and PCOS-NIR patients. Compared with the healthy control group, the upregulation of miR-93 decreased the expression of GLUT4 and impaired glucose transmembrane transport in AT of PCOS patients, which suggested that miR-93 plays a major part in PCOS-IR [59]. In addition, miR-93 potentially regulates signaling pathways of hepatic growth factor (HGF) detected in the ovary [103] and increases insulin secretion by increasing the amount of islet beta-cells through compensatory mechanisms and thus leads to IR. HGF is an adipokine and related to obesity and metabolic syndrome [104–106]. Another study found that miR-33b-5p targeted HMGA2 and SREBF1 to inhibit GLUT4 production and resulted in IR of PCOS [86]. We can speculate that certain miRNA might affect glucose metabolism and lead to IR in PCOS by regulating the expression of GLUT4.
Insulin-like growth factor 1 (IGF-1) is a peptide that is closely related to carbohydrate metabolic disturbance, including IR, type 2 diabetes, high blood pressure, obesity, and metabolic syndrome [107]. A study found that miR-122 mimic directly targeted the 3′ UTR of IGF-1 mRNA to significantly downregulate IGF-1 expression levels, suggesting that miR-122 can inhibit IGF-1 expression and decrease insulin sensitivity [108]. Four highly expressed genes (SOCS3 [109], SRF [110], FOXO1 [111], and PTEN [112]) were targeted by miR-483-5p, which is associated with IR and glucose intolerance in CCs of PCOS patients [77]. In addition, miR-485-3p can target peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) [87], which is a mediator of insulin signaling and maintains the function and integrity of mitochondria [113] and has lower expression levels in PCOS women [88]. The underlying mechanism is still unclear.
miRNAs and lipid metabolism in PCOS-IR
Besides endogenous IR, about 70% of PCOS patients have weight-related IR with dysfunctional and impaired glucose metabolism of AT. The lipid level can increase with the increase of glucose concentration in PCOS patients [114]. Legro et al. reported that the incidence of IR in PCOS obese patients was significantly higher than that in PCOS non-obese patients [115]. Adipocytes with IR have differentially expressed miRNAs [116], and several miRNAs are related to carbohydrate metabolism in AT. Diaz et al. found that miR-451a was the lowest level of miRNAs in PCOS compared with healthy people [90] and suppressed hepatic gluconeogenesis by downregulating glycerin kinase [117], which is considered a key driver of PCOS. It may target calcium-binding protein 39 (CAB39) to interfere with the PI3K/AKT signaling pathway in vivo [118] and lead to the suppression of the PI3K/AKT/mTOR pathway that is associated with diabetes, obesity, high blood pressure, and cancer [119–121]. The overexpression of miR-451a can also reduce triglyceride accumulation in mouse liver and cultured hepatocytes [122]. However, there is no sufficient evidence illustrating the function of miR-451a in PCOS-IR at present, which indicates a new research direction. Research showed that AT-specific GLUT4 could regulate tissue glucose tolerance, insulin sensitivity, and systemic glucose metabolism [101, 102]. In 3T3-L1 adipocytes, along with the increased expression levels of miR-320, IR was induced with high levels of glucose and insulin, and when adding anti-miR-320 oligos, GLUT4 expression was upregulated, and IR was brought back to normal [67]. Since miR-320 is abundant in FF of PCOS women [55], it can serve as a potential target to improve insulin sensitivity and IR. Although miR-223 can regulate the expression of GLUT4 in cardiomyocytes [93], there is no significant association among miR-223, GLUT4, and the evaluation of homeostasis model assessment of IR (HOMA-IR) in PCOS-AT [59]. When compared to the control group, miR-146a and miR-155 are shown to be differently expressed in GCs from the PCOS women [123]. miR-146a is involved in IR with higher expression under inflammatory stimulation in type 2 diabetic patients [124, 125], indicating the overexpressed miR-146a may contribute to inflammation and aggravate IR in PCOS. Gonzalez et al. found that a pro-oxidative and pro-inflammatory state caused by saturated fat intake may be an important factor for IR in PCOS [126]. miR-155 takes effect in autoimmune diseases and chronic inflammatory diseases [127–129] and regulates insulin sensitivity in both in vivo and in vitro mouse models [130, 131]. In addition, a study showed that the combined application of miR-193b and BMI might potentially be a new indicator to predict the incidence of impaired glucose metabolism in patients with PCOS [46]. More strong evidence demonstrating this relationship is that metformin can normalize the phenotype of PCOS more than oral contraceptive (OC) by reducing hepato-visceral adiposity and inducing IR [132]. However, no research has looked into the change in miRNA expression after metformin treatment among PCOS patients [133].
miRNAs and hyperandrogenemia in PCOS-IR
Hyperandrogenemia is a prominent pathological feature and clinical manifestation of PCOS and characterized by increased testosterone levels, which is associated with the abnormal distribution of abdominal fat [134], IR [135], and ovulatory dysfunction [136] in PCOS. Insulin stimulates androgen production by activating 17-hydroxylase/C17-20 lyase cytochrome P50 (CYP17) enzyme, decreasing the production of SHBG, increasing cAMP concentrations by working in coordination with LH, and resulting in StAR activation and androgen production [137, 138]. Hyperinsulinemia may lead to the onset of hyperandrogenemia in PCOS. In FF among PCOS patients, a study provided evidence that the miRNA expression profile was altered in three groups (PCOS-normo-androgenic, PCOS-hyperandrogenic, and healthy group) and specific miRNAs were associated with phenotypical traits of PCOS [27]. Further research showed that 750 miRNAs were differentially expressed in the three groups using TaqMan low-density arrays, and hyperandrogenic PCOS women showed more severe IR compared to normo-androgenic PCOS women and had a higher prevalence of metabolic syndrome [57]. In ovarian theca cells, a study on the miRNA profile in PCOS ovarian theca tissue showed that miR-92a might target IRS-2 and GATA6 [36] indicating cross-talk relationships between androgenic and insulin signaling pathways. However, in a case-control study, PCOS patients with elevated insulin and testosterone had higher levels of miR-93 and miR-223 in circulation, which were independent of IR or HA when compared to age- and weight-matched controls [60]. miRNAs in PCOS-IR and hyperandrogenemia might provide a new direction to understand the pathology and thus find potential therapeutic targets but requires further study.
miRNAs and endometrial receptivity in PCOS-IR
Among patients who undertake ART treatment, up to 67% with recurrent implantation failure is associated with poor endometrial receptivity (ER), which is closely regulated by multiple genes, signal transduction pathways, and cell-cell interactions [139]. A study showed that oocytes matured in vitro from PCOS-IR did not affect oocyte and embryo quality but did lower the pregnancy rate; thus, we may infer that IR and hyperinsulinemia can injure endometrial receptivity and disturb the embryo implantation process [140]. Romina et al. found that the expression of IRS-1, IRS-2, and the degree of tyrosine phosphorylation in endometrial tissues of PCOS-IR patients were lower than those of PCOS-NIR patients, suggesting that insulin signaling molecules may alter downstream kinase activity and trigger endometrial IR [141]. Compared with PCOS-NIR patients, cortisol was increased and inhibited the phosphorylation of protein kinase B (AKT) and GLUT-4 translocation by inducing phosphatase and leading to PCOS-IR in the endometrium [142]. Furthermore, ER chips showed that miRNAs could mediate multiple genes to complete the recognition at the maternal-fetal interface [143], for example, miR-323-3p is supposed to be a critical region for placental growth and embryonic development [144]. There are 44 differentially expressed miRNAs of endometrial stromal cells in vitro decidualization [145], and several differentially expressed miRNAs were associated with endometrial status in healthy women in the pre-implantation and implantation phases, late proliferate phase, and mid-secretion phases [146] in a previous study. Specifically, 148 miRNAs were differentially expressed in the uterus from the PCOS-IR rat model compared with the control [147]. Bioinformatic analysis and pathway analysis showed four miRNAs (− 484, − 375-3p, − 324-5p, and − 223-3p) and their target genes were involved in insulin secretion and signaling pathways, such as Wnt, AMPK, PI3K-Akt, and Ras. IGF and its related proteins were confirmed to participate in the physiological process of the endometrium by increasing apical fibronectin on blastocysts to increase attachment and invasion in an in vitro model of implantation [148]. Decidual cells of primates can secret IGF and possibly affect embryo implantation in the invasion stage of trophoblast cells [149] by targeting the androgen or IGF1 signaling pathway [36]. Investigating the alteration of the miRNA profile provides a better understanding of the mechanisms for poor ER in PCOS-IR; however, most of the studies are based on animal models, and few studies on miRNAs were conducted in human subjects, suggesting a potential research direction.
miRNAs and oocyte quality in PCOS-IR
Recent studies indicated that the dysfunction and abnormal expression of miRNAs in PCOS could affect the physiological function of the ovary and might lead to ovulation dysfunction and reproductive endocrine abnormalities [52, 96]. High insulin concentration can induce the arrest of follicular growth and development [150, 151]. For example, insulin can stimulate the proliferation of buffalo GCs at concentrations of 50 ng/ml and above [152]. The expression of miR-320a is increased in PCOS tissues in an insulin-dependent manner, and its downregulation decreases cell viability and colony formation and leads the cell apoptosis [94]. When KGN cells were treated with insulin, downregulated miR-483 promoted IGF-1 expression and insulin-induced cell proliferation [76] by targeting the IGF1 3′ UTR directly. After insulin treatment, the expression of miR-27a-3p was decreased in GCs by mediating signal transducers and transcription activators STAT1 and STAT3. It is worth noting that the overexpression of miR-27a-3p in KGN cells can reduce cell proliferation and promote apoptosis [96].
Furthermore, increased apoptosis of GCs and CCs may affect follicular development by preventing the conversion of androgens to estrogen and changing hormone levels in follicles [153, 154]. On the one hand, the apoptosis and dysfunction in CCs may be the important contributors to multiple symptoms in patients with PCOS [155], including irregular menstruation, follicular hypoplasia, anovulation, and infertility [156]. CCs provide nutrition and growth regulators essential for oocytes and secrete ovarian steroids and produce growth hormone [157]. In a case-control study, Hu et al. found that a total of 59 known miRNAs and 617 genes were differentially expressed in CCs of PCOS-IR. The target genes of miRNAs were analyzed by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation and found to contribute to the etiology and pathophysiology of PCOS-IR [158]. Rap1b is a regulator of the MAPK pathway and was suppressed directly by miR-612 in CCs of PCOS-IR [158]. IGF-1 is an effective mitogenic factor involved in the growth of preantral follicles in PCOS [159] and is essential for the development of ovarian follicles [160]. IGF-1 could act upon autocrine to stimulate CCs replication and differentiation [161], and elevated IGF-1 activity is proposed as a mechanism leading to the increased activation and accelerated growth of early-stage follicles within PCOS ovaries [162]. miR-323-3p targets IGF-1 to inhibit steroid production and apoptosis of CCs which leads to PCOS and has been reported to be downregulated in the FF from embryos with low quality [163]. It can also regulate the expression of epidermal growth factor receptor (EGFR), a marker of mammalian oocyte capacity [164], and affect meiosis and maturation of oocytes [91]. Another miRNA, mir-99a, regulates the apoptosis and proliferation of human GCs in PCOS by targeting IGF-1R [165]. On the other hand, studies showed that GC dysfunction with reduced proliferation, increased apoptosis, or imbalanced hormone secretion [166, 167] is one of the causes of abnormal follicle formation in PCOS [168]. Proliferating GCs provide a suitable microenvironment for the initiation of follicular growth, oocyte maturation, and atresia [169], and the communication is essential for the maintenance of normal fertility [170]. Recent studies found the increased GC apoptosis in patients with PCOS and PCOS animal models [171, 172]. There was an increased high-mobility group box 1 (HMGB1) in the serum from PCOS-IR women compared to PCOS-NIR, which promotes apoptosis in primary cultured rat ovarian GCs [173], and miR-204 downregulation can induce HMGB1 upregulation, promote proliferation, and inhibit apoptosis of GCs [174]. A study showed increased miR-93 targeted CDKN1A, which promotes the proliferation and cell cycle progression of GCs in PCOS [175], and it may be explained by the high concentration of insulin in PCOS [89]. Of note, metformin can effectively restore the periodic and natural ovulation of patients with PCOS by increasing the expression of CDKN1A in cultured bovine GCs [176]. miR-370 is a scaffold protein involved in the insulin pathway modulating IRS-1 expression [81] and the downregulation of miR-370 may target IRS-1 leading to disturbed insulin metabolism of GCs and affected oocyte quality [82, 83]. However, there is no relevant research confirming this hypothesis. These related miRNAs may influence the oocyte quality in PCOS-IR and may contribute to the adverse outcomes in PCOS patients undergoing ART, but there have been only a few reports about it.
Common differentially expressed miRNAs in different phenotypes in PCOS-IR
We reviewed and summarized the miRNAs that were altered in PCOS-IR and found that some common miRNAs can target multiple metabolic pathways, including insulin metabolism, androgen metabolism, lipid metabolism, and oogenesis. We explored the possible mechanisms in the insulin signaling pathways of PCOS-IR (Fig. 1). The expression of miR-323-3p in the CCs was downregulated in the PCOS group and was inversely proportional to the expression level of IGF-1, which was the direct target of miR-323-3p demonstrated by the dual-luciferase reporter assay [91]. Qing et al. detected miR-323-3p in the micro-vesicles-free FF from PCOS and control patients [55]. Moreover, IGF-1 may play a significant role in the regulation of steroidogenesis and apoptosis of CCs in the process of follicular development via the PI3K pathway [177]. In addition, Yang et al. [178] reported that a higher expression of IGF-1 could increase the vulnerability of granulosa cells, which eventually led to cell apoptosis. These results provide evidence that miR-323-3p, along with its target IGF-1, regulated steroid metabolism and cell viability.
miR-99a expression was downregulated in PCOS, and insulin could lower miR-99a expression in a dose-dependent manner, especially at a level above 80 ng/ml [165], so we speculate that the hyperphysiological dose of insulin in PCOS patients might be one of the reasons for decreased miR-99a level in GCs. Li et al. found that insulin-induced IGF-1/mTOR pathway activation was accompanied by a significantly reduced miR-99a level, and elevated IGF-1 enhanced IR in patients with PCOS. This indicates that IR, IGF system, and the expression of miR-99a possess some latent mutual effects [179]. Apart from the influence on folliculogenesis, a high concentration of free serum IGF-1 in patients with PCOS can adversely affect embryo implantation, causing higher blastocyst absorption rate and pregnancy miscarriages [180, 181]. Therefore, the upregulation of miR-99a might be an effective method to attenuate the adverse effects of excessive activation of the IGF-1/IGF-1R system in PCOS. At present, many studies have recognized that miRNAs target several genes and metabolic pathways, leading to PCOS-IR. However, the specific mechanism is not well understood. It is our focus to analyze these essential miRNAs related to PCOS-IR.
Conclusion
PCOS is the most common endocrine disease in women at childbearing age [182], and there are 3 different diagnostic criteria currently: NIH criteria [183], Rotterdam criteria [184], and Androgen Excess and PCOS Society [185], which are yet to be unified. Considering that women with PCOS already have an aberrant metabolic condition in their early adulthood, it is necessary to stratify PCOS patients by their metabolic risk, independently from the PCOS phenotypes. IR is one of the major determinants in the pathology of metabolic abnormalities, reproductive disorders, and the occurrence of PCOS. Several studies have shown that treatment targeting IR in PCOS can largely alleviate obesity and hyperandrogenism [186, 187]. Improved insulin sensitivity may facilitate glucose metabolism control, androgen level reduction, ovulation rate improvement, and thus improve fertility. miRNAs participate in hyperinsulinemia, ovarian insulin sensitivity, inflammation, and oocyte quality in PCOS. Due to the specificity of miRNAs, they may become unique and reliable biomarkers for the diagnosis of PCOS and can be used to distinguish the different phenotypes as determined by the Rotterdam criteria.
However, at present, we are still unable to identify the exact targets of dysfunctional miRNAs and their direct downstream processes. For example, ovarian function and regulation are highly species specific, and several human-specific miRNAs have been described [188], which appear to be GC specific. Therefore, maybe miRNAs in specific cell types of the ovary exist but have not been described or studied, which also plays a role in ovarian hormonal metabolism and development of PCOS. We speculate that the reasons may be the following: (1) the expression profiles of miRNAs may be related to race. Ding et al. found that there were no overlaps in changes to miRNAs between previous studies [52, 55, 80] and theirs, which may be due to the race of subjects (Han vs. the Caucasus) [189]. Moreover, studies showed an opposite effect of different miRNAs in FF of PCOS in China and the USA, and this may relate to race/ethnicity or PCOS phenotype and differences methodological [52, 55]. Therefore, it is important to investigate miRNA profiles from different races in a larger study; (2) in the previous studies, the sources of collected samples are diverse and cover a wide spectrum, such as plasma, FF, GCs, and CCs. As a result, the miRNA expression profiles found are different yet overlapping, which may be explained by the diversity of samples, the limitation of sample size, and the complexity and heterogeneity of PCOS; (3) each miRNA can have numerous gene targets and the gene targets may be different based on cell types, systems, and organs including the ovary [190]. miRNAs can also be regulated by other paracrine/autocrine factors, which makes it a highly complex system [191, 192].
Nowadays, several molecular approaches are rapidly developing which facilitate the identification of genomic regions associated with pregnancy establishment and fertility, including functional genomic approaches, structural genomics and integrated analysis of the transcriptome, and epigenome (DNA methylation and histone modification) using high-throughput next-generation sequencing techniques. High-throughput profiling and sequencing of miRNAs can provide an all-around tool to investigate all kinds of signaling pathways, improve clinical management, and determine appropriate infertility treatment based on abnormal follicular development in PCOS. With the help of novel technologies and searchable databases (including ontology and biological information databases), miRNA can provide us with exquisite analysis, prevention, and management for reproductive diseases like PCOS. The top priority is to conduct large-scale replication studies to identify certain miRNAs that have strong modulation effects in PCOS. Considering miRNAs often exist in families, an increasingly important approach is to modulate the levels of entire miRNA families or co-regulated miRNAs together as well as separately to establish roles in the intact tissues or organs. We can also analyze the functional role of single miRNAs in the pathology of PCOS-IR and verify the function of a single miRNA both in vivo and in vitro. It is worth noting that many miRNAs have the strongest effects at the level of translation inhibition, and the study of mRNA expression arrays may have limited use to reveal the real impact of regulated miRNAs on the PCOS phenotype. Recently, many miRNA mimics, anti-miRNA oligonucleotides, and small interfering RNA therapeutics are being evaluated. Currently, there is no treatment to target the miRNAs in PCOS-IR directly, and studying related miRNAs may pose new research directions for the treatment of PCOS-IR in the future to utilize miRNAs as biomarkers.
Authors’ contributions
Yingliu Luo: Writing-original draft preparation and editing
Chenchen Cui: Writing-reviewing and editing
Xiao Han: Writing-reviewing, editing, and supervision
Qian Wang: Supervision
Cuilian Zhang: Supervision, project administration, and funding acquisition
Funding
This work was supported by the National Natural Science Foundation of China (81571407).
Compliance with ethical standards
Conflict of interest
The authors declare that they have no conflicts of interest.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Skiba MA, Islam RM, Bell RJ, Davis SR. Understanding variation in prevalence estimates of polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod Update. 2018;24(6):694–709. doi: 10.1093/humupd/dmy022. [DOI] [PubMed] [Google Scholar]
- 2.Stepto NK, Cassar S, Joham AE, Hutchison SK, Harrison CL, Goldstein RF, et al. Women with polycystic ovary syndrome have intrinsic insulin resistance on euglycaemic-hyperinsulaemic clamp. Hum Reprod (Oxford, England) 2013;28(3):777–784. doi: 10.1093/humrep/des463. [DOI] [PubMed] [Google Scholar]
- 3.Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, et al. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2013;98(12):4565–4592. doi: 10.1210/jc.2013-2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Toulis KA, Goulis DG, Mintziori G, Kintiraki E, Eukarpidis E, Mouratoglou SA, et al. Meta-analysis of cardiovascular disease risk markers in women with polycystic ovary syndrome. Hum Reprod Update. 2011;17(6):741–760. doi: 10.1093/humupd/dmr025. [DOI] [PubMed] [Google Scholar]
- 5.Reyes-Munoz E, Ortega-Gonzalez C, Martinez-Cruz N, Arce-Sanchez L, Estrada-Gutierrez G, Moran C, et al. Association of obesity and overweight with the prevalence of insulin resistance, pre-diabetes and clinical-biochemical characteristics among infertile Mexican women with polycystic ovary syndrome: a cross-sectional study. BMJ Open. 2016;6(7):e012107. doi: 10.1136/bmjopen-2016-012107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Azziz R. PCOS in 2015: New insights into the genetics of polycystic ovary syndrome. Nat Rev Endocrinol 2016;12(2):74–75. doi:10.1038/nrendo.2015.230. [DOI] [PubMed]
- 7.Escobar-Morreale HF. Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nat Rev Endocrinol. 2018;14(5):270–284. doi: 10.1038/nrendo.2018.24. [DOI] [PubMed] [Google Scholar]
- 8.Miller WP, Ravi S, Martin TD, Kimball SR, Dennis MD. Activation of the stress response kinase JNK (c-Jun N-terminal kinase) attenuates insulin action in retina through a p70S6K1-dependent mechanism. J Biol Chem. 2017;292(5):1591–1602. doi: 10.1074/jbc.M116.760868.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sirmans SM, Pate KA. Epidemiology, diagnosis, and management of polycystic ovary syndrome. Clin Epidemiol. 2013;6:1–13. doi: 10.2147/clep.S37559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cakir E, Topaloglu O, Colak Bozkurt N, Karbek Bayraktar B, Gungunes A, Sayki Arslan M, et al. Insulin-like growth factor 1, liver enzymes, and insulin resistance in patients with PCOS and hirsutism. Turkish J Med Sci. 2014;44(5):781–786. doi: 10.3906/sag-1303-80. [DOI] [PubMed] [Google Scholar]
- 11.Tosi F, Negri C, Brun E, Castello R, Faccini G, Bonora E, et al. Insulin enhances ACTH-stimulated androgen and glucocorticoid metabolism in hyperandrogenic women. Eur J Endocrinol. 2011;164(2):197–203. doi: 10.1530/eje-10-0782. [DOI] [PubMed] [Google Scholar]
- 12.Adashi EY, Hsueh AJ, Yen SS. Insulin enhancement of luteinizing hormone and follicle-stimulating hormone release by cultured pituitary cells. Endocrinology. 1981;108(4):1441–1449. doi: 10.1210/endo-108-4-1441. [DOI] [PubMed] [Google Scholar]
- 13.Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications. Endocr Rev. 2012;33(6):981–1030. doi: 10.1210/er.2011-1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pasquali R, Stener-Victorin E, Yildiz BO, Duleba AJ, Hoeger K, Mason H, et al. PCOS forum: research in polycystic ovary syndrome today and tomorrow. Clin Endocrinol. 2011;74(4):424–433. doi: 10.1111/j.1365-2265.2010.03956.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gayoso-Diz P, Otero-González A, Rodriguez-Alvarez MX, Gude F, García F, De Francisco A, et al. Insulin resistance (HOMA-IR) cut-off values and the metabolic syndrome in a general adult population: effect of gender and age: EPIRCE cross-sectional study. BMC Endocr Disord. 2013;13:47. doi: 10.1186/1472-6823-13-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lewandowski KC, Skowrońska-Jóźwiak E, Łukasiak K, Gałuszko K, Dukowicz A, Cedro M et al. How much insulin resistance in polycystic ovary syndrome? Comparison of HOMA-IR and insulin resistance (Belfiore) index models. Arch Med Sci 2019;15(3):613–618. doi:10.5114/aoms.2019.82672. [DOI] [PMC free article] [PubMed]
- 17.Shashaj B, Luciano R, Contoli B, Morino GS, Spreghini MR, Rustico C, et al. Reference ranges of HOMA-IR in normal-weight and obese young Caucasians. Acta Diabetol. 2016;53(2):251–260. doi: 10.1007/s00592-015-0782-4. [DOI] [PubMed] [Google Scholar]
- 18.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–297. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
- 19.Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, et al. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433(7027):769–773. doi: 10.1038/nature03315. [DOI] [PubMed] [Google Scholar]
- 20.Flynt AS, Lai EC. Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nat Rev Genet. 2008;9(11):831–842. doi: 10.1038/nrg2455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bu X, Zhang J, Tian F, Wang X, Wu L, Tian W. Value of diffusion-weighted magnetic resonance imaging combined with miR-18a level in predicting radiosensitivity of cervical cancer. Med Sci Monit Int Med J Exp Clin Res. 2018;24:7271–7278. doi: 10.12659/msm.910990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mari-Alexandre J, Barcelo-Molina M, Belmonte-Lopez E, Garcia-Oms J, Estelles A, Braza-Boils A et al. Micro-RNA profile and proteins in peritoneal fluid from women with endometriosis: their relationship with sterility. Fertil Steril. 2018;109(4):675–84.e2. doi:10.1016/j.fertnstert.2017.11.036. [DOI] [PubMed]
- 23.Romakina VV, Zhirov IV, Nasonova SN, Zaseeva AV, Kochetov AG, Liang OV, et al. MicroRNAs as biomarkers of cardiovascular diseases. Kardiologiia. 2018;(1):66–71. 10.18087/cardio.2018.1.10083. [DOI] [PubMed]
- 24.Cortez MA, Bueso-Ramos C, Ferdin J, Lopez-Berestein G, Sood AK, Calin GA. MicroRNAs in body fluids--the mix of hormones and biomarkers. Nat Rev Clin Oncol. 2011;8(8):467–477. doi: 10.1038/nrclinonc.2011.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dumas ME, Emanueli C. Circulating microRNAs to predict the risk for metabolic diseases in the general population? Diabetes. 2017;66(3):565–567. doi: 10.2337/dbi16-0072. [DOI] [PubMed] [Google Scholar]
- 26.Guay C, Regazzi R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nat Rev Endocrinol. 2013;9(9):513–521. doi: 10.1038/nrendo.2013.86. [DOI] [PubMed] [Google Scholar]
- 27.Sorensen AE, Udesen PB, Wissing ML, Englund ALM, Dalgaard LT. MicroRNAs related to androgen metabolism and polycystic ovary syndrome. Chem Biol Interact. 2016;259(Pt A):8–16. doi: 10.1016/j.cbi.2016.06.008. [DOI] [PubMed] [Google Scholar]
- 28.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(5):1304–1315. doi:10.1159/000358698. [DOI] [PubMed]
- 29.Wu HL, Heneidi S, Chuang TY, Diamond MP, Layman LC, Azziz R, et al. The expression of the miR-25/93/106b family of micro-RNAs in the adipose tissue of women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2014;99(12):E2754–E2761. doi: 10.1210/jc.2013-4435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.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. doi: 10.1016/j.mce.2015.01.030. [DOI] [PubMed] [Google Scholar]
- 31.Naji M, Aleyasin A, Nekoonam S, Arefian E, Mahdian R, Amidi F. Differential expression of miR-93 and miR-21 in granulosa cells and follicular fluid of polycystic ovary syndrome associating with different phenotypes. Sci Rep. 2017;7(1):14671. doi: 10.1038/s41598-017-13250-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kamalidehghan B, Habibi M, Afjeh SS, Shoai M, Alidoost S, Almasi Ghale R, et al. The importance of small non-coding RNAs in human reproduction: a review article. Appl Clin Genet. 2020;13:1–11. doi: 10.2147/tacg.S207491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sørensen AE, Wissing ML, Salö S, Englund AL, Dalgaard LT. MicroRNAs related to polycystic ovary syndrome (PCOS) Genes. 2014;5(3):684–708. doi: 10.3390/genes5030684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chen B, Xu P, Wang J, Zhang C. The role of MiRNA in polycystic ovary syndrome (PCOS) Gene. 2019;706:91–96. doi: 10.1016/j.gene.2019.04.082. [DOI] [PubMed] [Google Scholar]
- 35.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(8):754–762. doi: 10.1089/dna.2019.4622. [DOI] [PubMed] [Google Scholar]
- 36.Lin L, Du T, Huang J, Huang LL, Yang DZ. Identification of differentially expressed microRNAs in the ovary of polycystic ovary syndrome with hyperandrogenism and insulin resistance. Chin Med J. 2015;128(2):169–174. doi: 10.4103/0366-6999.149189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Belani M, Deo A, Shah P, Banker M, Singal P, Gupta S. Differential insulin and steroidogenic signaling in insulin resistant and non-insulin resistant human luteinized granulosa cells-a study in PCOS patients. J Steroid Biochem Mol Biol. 2018;178:283–292. doi: 10.1016/j.jsbmb.2018.01.008. [DOI] [PubMed] [Google Scholar]
- 38.Cakiroglu Y, Doger E, Vural F, Kopuk SY, Vural B. Impact of insulin resistance and obesity on intracytoplasmic sperm injection outcomes in young women with polycystıc ovary syndrome. North Clin Istanbul. 2017;4(3):218–224. doi: 10.14744/nci.2017.79663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Macut D, Bjekic-Macut J, Rahelic D, Doknic M. Insulin and the polycystic ovary syndrome. Diabetes Res Clin Pract. 2017;130:163–170. doi: 10.1016/j.diabres.2017.06.011. [DOI] [PubMed] [Google Scholar]
- 40.Grasedieck S, Schöler N, Bommer M, Niess JH, Tumani H, Rouhi A, et al. Impact of serum storage conditions on microRNA stability. Leukemia. 2012;26(11):2414–2416. doi: 10.1038/leu.2012.106. [DOI] [PubMed] [Google Scholar]
- 41.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(10):997–1006. doi: 10.1038/cr.2008.282. [DOI] [PubMed] [Google Scholar]
- 42.Ortega FJ, Mercader JM, Moreno-Navarrete JM, Rovira O, Guerra E, Esteve E, et al. Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization. Diabetes Care. 2014;37(5):1375–1383. doi: 10.2337/dc13-1847. [DOI] [PubMed] [Google Scholar]
- 43.Sirotkin AV, Ovcharenko D, Grossmann R, Lauková M, Mlyncek M. Identification of microRNAs controlling human ovarian cell steroidogenesis via a genome-scale screen. J Cell Physiol. 2009;219(2):415–420. doi: 10.1002/jcp.21689. [DOI] [PubMed] [Google Scholar]
- 44.Nanda D, Chandrasekaran SP, Ramachandran V, Kalaivanan K, Carani Venkatraman A. Evaluation of serum miRNA-24, miRNA-29a and miRNA-502-3p expression in PCOS subjects: correlation with biochemical parameters related to PCOS and insulin resistance. Indian J Clin Biochem. 2020;35(2):169–178. doi: 10.1007/s12291-018-0808-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Melkman-Zehavi T, Oren R, Kredo-Russo S, Shapira T, Mandelbaum AD, Rivkin N, et al. miRNAs control insulin content in pancreatic β-cells via downregulation of transcriptional repressors. EMBO J. 2011;30(5):835–845. doi: 10.1038/emboj.2010.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Jiang L, Huang J, Chen Y, Yang Y, Li R, Li Y, et al. Identification of several circulating microRNAs from a genome-wide circulating microRNA expression profile as potential biomarkers for impaired glucose metabolism in polycystic ovarian syndrome. Endocrine. 2016;53(1):280–290. doi: 10.1007/s12020-016-0878-9. [DOI] [PubMed] [Google Scholar]
- 47.Wang R, Hong J, Cao Y, Shi J, Gu W, Ning G, et al. Elevated circulating microRNA-122 is associated with obesity and insulin resistance in young adults. Eur J Endocrinol. 2015;172(3):291–300. doi: 10.1530/eje-14-0867. [DOI] [PubMed] [Google Scholar]
- 48.Jiang B, Xue M, Xu D, Song Y, Zhu S. Upregulation of microRNA-204 improves insulin resistance of polycystic ovarian syndrome via inhibition of HMGB1 and the inactivation of the TLR4/NF-kappaB pathway. Cell Cycle (Georgetown, Tex). 2020:1–14. 10.1080/15384101.2020.1724601. [DOI] [PMC free article] [PubMed] [Retracted]
- 49.Shuai F, Wang B, Dong S. MicroRNA-204 inhibits the growth and motility of colorectal cancer cells by downregulation of CXCL8. Oncol Res. 2018;26(8):1295–1305. doi: 10.3727/096504018x15172747209020. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 50.Da Broi MG, Giorgi VSI, Wang F, Keefe DL, Albertini D, Navarro PA. Influence of follicular fluid and cumulus cells on oocyte quality: clinical implications. J Assist Reprod Genet. 2018;35(5):735–751. doi: 10.1007/s10815-018-1143-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dai G, Lu G. Different protein expression patterns associated with polycystic ovary syndrome in human follicular fluid during controlled ovarian hyperstimulation. Reprod Fertil Dev. 2012;24(7):893–904. doi: 10.1071/rd11201. [DOI] [PubMed] [Google Scholar]
- 52.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(3):355–362. doi: 10.1007/s10815-013-0161-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Tucker WC, Chapman ER. Role of synaptotagmin in Ca2+-triggered exocytosis. Biochem J. 2002;366(Pt 1):1–13. doi: 10.1042/bj20020776.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lang J, Fukuda M, Zhang H, Mikoshiba K, Wollheim CB. The first C2 domain of synaptotagmin is required for exocytosis of insulin from pancreatic beta-cells: action of synaptotagmin at low micromolar calcium. EMBO J. 1997;16(19):5837–5846. doi: 10.1093/emboj/16.19.5837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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(7):3068–3079. doi: 10.1210/jc.2013-1715. [DOI] [PubMed] [Google Scholar]
- 56.Lamadrid-Romero M, Solis KH, Cruz-Resendiz MS, Perez JE, Diaz NF, Flores-Herrera H, et al. Central nervous system development-related microRNAs levels increase in the serum of gestational diabetic women during the first trimester of pregnancy. Neurosci Res. 2018;130:8–22. doi: 10.1016/j.neures.2017.08.003. [DOI] [PubMed] [Google Scholar]
- 57.Sorensen AE, Udesen PB, Maciag G, Geiger J, Saliani N, Januszewski AS, et al. Hyperandrogenism and metabolic syndrome are associated with changes in serum-derived microRNAs in women with polycystic ovary syndrome. Front Med. 2019;6:242. doi: 10.3389/fmed.2019.00242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yin M, Wang X, Yao G, Lü M, Liang M, Sun Y, et al. 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. doi: 10.1074/jbc.M113.546044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.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(7):2278–2286. doi: 10.2337/db12-0963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.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. doi: 10.1038/srep16890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ho CK, Wood JR, Stewart DR, Ewens K, Ankener W, Wickenheisser J, et al. Increased transcription and increased messenger ribonucleic acid (mRNA) stability contribute to increased GATA6 mRNA abundance in polycystic ovary syndrome theca cells. J Clin Endocrinol Metab. 2005;90(12):6596–6602. doi: 10.1210/jc.2005-0890. [DOI] [PubMed] [Google Scholar]
- 62.Zhang C, Yu C, Lin Z, Pan H, Li K, Ma H. MiRNAs expression profiling of rat ovaries displaying PCOS with insulin resistance. Arch Gynecol Obstet. 2020. 10.1007/s00404-020-05730-z. [DOI] [PMC free article] [PubMed]
- 63.Motta AB. Epigenetic marks in polycystic ovary syndrome. Curr Med Chem. 2019. 10.2174/0929867326666191003154548. [DOI] [PubMed]
- 64.Chen C, Zhao Z, Liu Y, Mu D. microRNA-99a is downregulated and promotes proliferation, migration and invasion in non-small cell lung cancer A549 and H1299 cells. Oncol Lett. 2015;9(3):1128–1134. doi: 10.3892/ol.2015.2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.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. doi: 10.1038/srep24976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.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(6):810–817. doi: 10.1161/circresaha.110.226357. [DOI] [PubMed] [Google Scholar]
- 67.Ling HY, Ou HS, Feng SD, Zhang XY, Tuo QH, Chen LX, et al. CHANGES IN microRNA (miR) profile and effects of miR-320 in insulin-resistant 3T3-L1 adipocytes. Clin Exp Pharmacol Physiol. 2009;36(9):e32–e39. doi: 10.1111/j.1440-1681.2009.05207.x. [DOI] [PubMed] [Google Scholar]
- 68.Hamdan M, Dunselman G, Li TC, Cheong Y. The impact of endometrioma on IVF/ICSI outcomes: a systematic review and meta-analysis. Hum Reprod Update. 2015;21(6):809–825. doi: 10.1093/humupd/dmv035. [DOI] [PubMed] [Google Scholar]
- 69.Surrey ES. Endometriosis-related infertility: the role of the assisted reproductive technologies. Biomed Res Int. 2015;2015:482959. doi: 10.1155/2015/482959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.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 Ovar Res. 2019;12(1):39. doi: 10.1186/s13048-019-0513-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.El-Shal AS, Zidan HE, Rashad NM, Abdelaziz AM, Harira MM. Association between genes encoding components of the leutinizing hormone/luteinizing hormone-choriogonadotrophin receptor pathway and polycystic ovary syndrome in Egyptian women. IUBMB Life. 2016;68(1):23–36. doi: 10.1002/iub.1457. [DOI] [PubMed] [Google Scholar]
- 72.Zhao C, Dong J, Jiang T, Shi Z, Yu B, Zhu Y, et al. Early second-trimester serum miRNA profiling predicts gestational diabetes mellitus. PLoS One. 2011;6(8):e23925. doi: 10.1371/journal.pone.0023925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.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(1):221–231. doi: 10.1007/s00404-017-4570-y. [DOI] [PubMed] [Google Scholar]
- 74.Yan G, Zhang L, Fang T, Zhang Q, Wu S, Jiang Y, et al. MicroRNA-145 suppresses mouse granulosa cell proliferation by targeting activin receptor IB. FEBS Lett. 2012;586(19):3263–3270. doi: 10.1016/j.febslet.2012.06.048. [DOI] [PubMed] [Google Scholar]
- 75.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 (Thousand Oaks, Calif). 2017;24(6):902–910. doi:10.1177/1933719116673197. [DOI] [PubMed]
- 76.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;22:3383–3393. doi: 10.12659/msm.897301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Shi L, Liu S, Zhao W, Shi J. miR-483-5p and miR-486-5p are down-regulated in cumulus cells of metaphase II oocytes from women with polycystic ovary syndrome. Reprod BioMed Online. 2015;31(4):565–572. doi: 10.1016/j.rbmo.2015.06.023. [DOI] [PubMed] [Google Scholar]
- 78.Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007;129(7):1401–1414. doi: 10.1016/j.cell.2007.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ma N, Wang X, Qiao Y, Li F, Hui Y, Zou C, et al. Coexpression of an intronic microRNA and its host gene reveals a potential role for miR-483-5p as an IGF2 partner. Mol Cell Endocrinol. 2011;333(1):96–101. doi: 10.1016/j.mce.2010.11.027. [DOI] [PubMed] [Google Scholar]
- 80.Murri M, Insenser M, Fernandez-Duran E, San-Millan 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(11):E1835–E1844. doi: 10.1210/jc.2013-2218. [DOI] [PubMed] [Google Scholar]
- 81.Fan C, Liu S, Zhao Y, Han Y, Yang L, Tao G, et al. Upregulation of miR-370 contributes to the progression of gastric carcinoma via suppression of FOXO1. Biomed Pharmacother = Biomedecine & pharmacotherapie. 2013;67(6):521–526. doi: 10.1016/j.biopha.2013.04.014. [DOI] [PubMed] [Google Scholar]
- 82.Chang KW, Chu TH, Gong NR, Chiang WF, Yang CC, Liu CJ, et al. miR-370 modulates insulin receptor substrate-1 expression and inhibits the tumor phenotypes of oral carcinoma. Oral Dis. 2013;19(6):611–619. doi: 10.1111/odi.12046. [DOI] [PubMed] [Google Scholar]
- 83.Fontes G, Ghislain J, Benterki I, Zarrouki B, Trudel D, Berthiaume Y et al. The deltaF508 mutation in the cystic fibrosis transmembrane conductance regulator is associated with progressive insulin resistance and decreased functional beta-cell mass in mice. Diabetes. 2015;64(12):4112–4122. doi:10.2337/db14-0810. [DOI] [PMC free article] [PubMed]
- 84.Hu MH, Zheng SX, Yin H, Zhu XY, Lu FT, Tong XH et al. Identification of microRNAs that regulate the MAPK pathway in human cumulus cells from PCOS women with insulin resistance. Reprod Sci (Thousand Oaks, Calif). 2020. doi:10.1007/s43032-019-00086-5. [DOI] [PubMed]
- 85.Chuang TY, Wu HL, Chen CC, Gamboa GM, Layman LC, Diamond MP, et al. MicroRNA-223 expression is upregulated in insulin resistant human adipose tissue. J Diabetes Res. 2015;2015:943659. doi: 10.1155/2015/943659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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(6):3073–3085. doi: 10.3892/or.2018.6375. [DOI] [PubMed] [Google Scholar]
- 87.Lou C, Xiao M, Cheng S, Lu X, Jia S, Ren Y, et al. MiR-485-3p and miR-485-5p suppress breast cancer cell metastasis by inhibiting PGC-1alpha expression. Cell Death Dis. 2016;7:e2159. doi: 10.1038/cddis.2016.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Skov V, Glintborg D, Knudsen S, Jensen T, Kruse TA, Tan Q, et al. Reduced expression of nuclear-encoded genes involved in mitochondrial oxidative metabolism in skeletal muscle of insulin-resistant women with polycystic ovary syndrome. Diabetes. 2007;56(9):2349–2355. doi: 10.2337/db07-0275. [DOI] [PubMed] [Google Scholar]
- 89.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(5):E729–E738. doi: 10.1210/jc.2014-3827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Diaz M, Bassols J, Lopez-Bermejo A, de Zegher F, Ibanez L. Low circulating levels of miR-451a in girls with polycystic ovary syndrome: different effects of randomized treatments. J Clin Endocrinol Metab. 2020;105(3). 10.1210/clinem/dgz204. [DOI] [PubMed]
- 91.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. doi:10.1016/j.gene.2018.10.006. [DOI] [PubMed]
- 92.Zhang T, Duan J, Zhang L, Li Z, Steer CJ, Yan G et al. LXRalpha promotes hepatosteatosis in part through activation of microRNA-378 transcription and inhibition of Ppargc1beta expression. Hepatology (Baltimore, Md). 2019;69(4):1488–1503. doi:10.1002/hep.30301. [DOI] [PMC free article] [PubMed]
- 93.Qin L, Chen J, Tang L, Zuo T, Chen H, Gao R, et al. Significant role of Dicer and miR-223 in adipose tissue of polycystic ovary syndrome patients. Biomed Res Int. 2019;2019:9193236. doi: 10.1155/2019/9193236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yang L, Li Y, Wang X, Liu Y, Yang L. MicroRNA320a inhibition decreases insulininduced KGN cell proliferation and apoptosis by targeting PCGF1. Mol Med Rep. 2017;16(4):5706–5712. doi: 10.3892/mmr.2017.7270. [DOI] [PubMed] [Google Scholar]
- 95.Yu Y, Chai J, Zhang H, Chu W, Liu L, Ma L et al. miR-194 promotes burn-induced hyperglycemia via attenuating IGF-IR expression. Shock (Augusta, Ga). 2014;42(6):578–584. doi:10.1097/shk.0000000000000258. [DOI] [PubMed]
- 96.Wang M, Sun J, Xu B, Chrusciel M, Gao J, Bazert M, et al. Functional characterization of microRNA-27a-3p expression in human polycystic ovary syndrome. Endocrinology. 2018;159(1):297–309. doi: 10.1210/en.2017-00219. [DOI] [PubMed] [Google Scholar]
- 97.Xiong Z, Li B, Wang W, Zeng X, Li B, Jian S, et al. MiR-140 targets RAP2A to enable the proliferation of insulin-treated ovarian granulosa cells. J Ovarian Res. 2020;13(1):13. doi: 10.1186/s13048-020-0611-4. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 98.Ding Y, He P, Li Z. MicroRNA-9119 regulates cell viability of granulosa cells in polycystic ovarian syndrome via mediating Dicer expression. Mol Cell Biochem. 2020;465(1–2):187–197. doi: 10.1007/s11010-019-03678-6. [DOI] [PubMed] [Google Scholar]
- 99.Chakraborty C, Roy SS, Hsu MJ, Agoramoorthy G. Landscape mapping of functional proteins in insulin signal transduction and insulin resistance: a network-based protein-protein interaction analysis. PLoS One. 2011;6(1):e16388. doi: 10.1371/journal.pone.0016388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Thomson MJ, Williams MG, Frost SC. Development of insulin resistance in 3T3-L1 adipocytes. J Biol Chem. 1997;272(12):7759–7764. doi: 10.1074/jbc.272.12.7759. [DOI] [PubMed] [Google Scholar]
- 101.Abel ED, Peroni O, Kim JK, Kim YB, Boss O, Hadro E, et al. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature. 2001;409(6821):729–733. doi: 10.1038/35055575. [DOI] [PubMed] [Google Scholar]
- 102.Tozzo E, Shepherd PR, Gnudi L, Kahn BB. Transgenic GLUT-4 overexpression in fat enhances glucose metabolism: preferential effect on fatty acid synthesis. Am J Phys. 1995;268(5 Pt 1):E956–E964. doi: 10.1152/ajpendo.1995.268.5.E956. [DOI] [PubMed] [Google Scholar]
- 103.Sahin N, Toylu A, Gulekli B, Dogan E, Kovali M, Atabey N. The levels of hepatocyte growth factor in serum and follicular fluid and the expression of c-Met in granulosa cells in patients with polycystic ovary syndrome. Fertil Steril. 2013;99(1):264–269. doi: 10.1016/j.fertnstert.2012.08.059. [DOI] [PubMed] [Google Scholar]
- 104.Vistoropsky Y, Trofimov S, Malkin I, Kobyliansky E, Livshits G. Genetic and environmental determinants of hepatocyte growth factor levels and their association with obesity and blood pressure. Ann Hum Biol. 2008;35(1):93–103. doi: 10.1080/03014460701822003. [DOI] [PubMed] [Google Scholar]
- 105.Araujo TG, Oliveira AG, Carvalho BM, Guadagnini D, Protzek AO, Carvalheira JB, et al. Hepatocyte growth factor plays a key role in insulin resistance-associated compensatory mechanisms. Endocrinology. 2012;153(12):5760–5769. doi: 10.1210/en.2012-1496. [DOI] [PubMed] [Google Scholar]
- 106.Hiratsuka A, Adachi H, Fujiura Y, Yamagishi S, Hirai Y, Enomoto M, et al. Strong association between serum hepatocyte growth factor and metabolic syndrome. J Clin Endocrinol Metab. 2005;90(5):2927–2931. doi: 10.1210/jc.2004-1588. [DOI] [PubMed] [Google Scholar]
- 107.Djiogue S, Nwabo Kamdje AH, Vecchio L, Kipanyula MJ, Farahna M, Aldebasi Y, et al. Insulin resistance and cancer: the role of insulin and IGFs. Endocr Relat Cancer. 2013;20(1):R1–r17. doi: 10.1530/erc-12-0324. [DOI] [PubMed] [Google Scholar]
- 108.Dong L, Hou X, Liu F, Tao H, Zhang Y, Zhao H, et al. Regulation of insulin resistance by targeting the insulin-like growth factor 1 receptor with microRNA-122-5p in hepatic cells. Cell Biol Int. 2019;43(5):553–564. doi: 10.1002/cbin.11129. [DOI] [PubMed] [Google Scholar]
- 109.Dey BR, Furlanetto RW, Nissley P. Suppressor of cytokine signaling (SOCS)-3 protein interacts with the insulin-like growth factor-I receptor. Biochem Biophys Res Commun. 2000;278(1):38–43. doi: 10.1006/bbrc.2000.3762. [DOI] [PubMed] [Google Scholar]
- 110.Jin W, Goldfine AB, Boes T, Henry RR, Ciaraldi TP, Kim EY, et al. Increased SRF transcriptional activity in human and mouse skeletal muscle is a signature of insulin resistance. J Clin Invest. 2011;121(3):918–929. doi: 10.1172/jci41940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kousteni S. FoxO1, the transcriptional chief of staff of energy metabolism. Bone. 2012;50(2):437–443. doi: 10.1016/j.bone.2011.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Wijesekara N, Konrad D, Eweida M, Jefferies C, Liadis N, Giacca A, et al. Muscle-specific Pten deletion protects against insulin resistance and diabetes. Mol Cell Biol. 2005;25(3):1135–1145. doi: 10.1128/mcb.25.3.1135-1145.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Pagel-Langenickel I, Bao J, Joseph JJ, Schwartz DR, Mantell BS, Xu X, et al. PGC-1alpha integrates insulin signaling, mitochondrial regulation, and bioenergetic function in skeletal muscle. J Biol Chem. 2008;283(33):22464–22472. doi: 10.1074/jbc.M800842200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wild RA. Dyslipidemia in PCOS. Steroids. 2012;77(4):295–299. doi: 10.1016/j.steroids.2011.12.002. [DOI] [PubMed] [Google Scholar]
- 115.Legro RS, Castracane VD, Kauffman RP. Detecting insulin resistance in polycystic ovary syndrome: purposes and pitfalls. Obstet Gynecol Surv. 2004;59(2):141–154. doi: 10.1097/01.Ogx.0000109523.25076.E2. [DOI] [PubMed] [Google Scholar]
- 116.Herrera BM, Lockstone HE, Taylor JM, Ria M, Barrett A, Collins S, et al. Global microRNA expression profiles in insulin target tissues in a spontaneous rat model of type 2 diabetes. Diabetologia. 2010;53(6):1099–1109. doi: 10.1007/s00125-010-1667-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Zhuo S, Yang M, Zhao Y, Chen X, Zhang F, Li N, et al. MicroRNA-451 negatively regulates hepatic glucose production and glucose homeostasis by targeting glycerol kinase-mediated gluconeogenesis. Diabetes. 2016;65(11):3276–3288. doi: 10.2337/db16-0166. [DOI] [PubMed] [Google Scholar]
- 118.Tian Y, Nan Y, Han L, Zhang A, Wang G, Jia Z et al. MicroRNA miR-451 downregulates the PI3K/AKT pathway through CAB39 in human glioma. Int J Oncol. 2012;40(4):1105–12. doi:10.3892/ijo.2011.1306. [DOI] [PMC free article] [PubMed]
- 119.Riquelme I, Tapia O, Leal P, Sandoval A, Varga MG, Letelier P, et al. miR-101-2, miR-125b-2 and miR-451a act as potential tumor suppressors in gastric cancer through regulation of the PI3K/AKT/mTOR pathway. Cell Oncol (Dordrecht) 2016;39(1):23–33. doi: 10.1007/s13402-015-0247-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Minna E, Romeo P, Dugo M, De Cecco L, Todoerti K, Pilotti S, et al. miR-451a is underexpressed and targets AKT/mTOR pathway in papillary thyroid carcinoma. Oncotarget. 2016;7(11):12731–12747. doi: 10.18632/oncotarget.7262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ong PS, Wang LZ, Dai X, Tseng SH, Loo SJ, Sethi G. Judicious toggling of mTOR activity to combat insulin resistance and cancer: current evidence and perspectives. Front Pharmacol. 2016;7:395. doi: 10.3389/fphar.2016.00395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Zeng N, Huang R, Li N, Jiang H, Li R, Wang F, et al. MiR-451a attenuates free fatty acids-mediated hepatocyte steatosis by targeting the thyroid hormone responsive spot 14 gene. Mol Cell Endocrinol. 2018;474:260–271. doi: 10.1016/j.mce.2018.03.016. [DOI] [PubMed] [Google Scholar]
- 123.Cirillo F, Catellani C, Lazzeroni P, Sartori C, Nicoli A, Amarri S, et al. MiRNAs regulating insulin sensitivity are dysregulated in polycystic ovary syndrome (PCOS) ovaries and are associated with markers of inflammation and insulin sensitivity. Front Endocrinol. 2019;10:879. doi: 10.3389/fendo.2019.00879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Duan Q, Mao X, Xiao Y, Liu Z, Wang Y, Zhou H, et al. Super enhancers at the miR-146a and miR-155 genes contribute to self-regulation of inflammation. Biochim Biophys Acta. 2016;1859(4):564–571. doi: 10.1016/j.bbagrm.2016.02.004. [DOI] [PubMed] [Google Scholar]
- 125.Balasubramanyam M, Aravind S, Gokulakrishnan K, Prabu P, Sathishkumar C, Ranjani H, et al. Impaired miR-146a expression links subclinical inflammation and insulin resistance in type 2 diabetes. Mol Cell Biochem. 2011;351(1–2):197–205. doi: 10.1007/s11010-011-0727-3. [DOI] [PubMed] [Google Scholar]
- 126.Gonzalez F, Considine RV, Abdelhadi OA, Acton AJ. Inflammation triggered by saturated fat ingestion is linked to insulin resistance and hyperandrogenism in PCOS. J Clin Endocrinol Metab. 2020. 10.1210/clinem/dgaa108. [DOI] [PMC free article] [PubMed]
- 127.Alivernini S, Gremese E, McSharry C, Tolusso B, Ferraccioli G, McInnes IB et al. MicroRNA-155-at the critical interface of innate and adaptive immunity in arthritis. Front Immunol 2017;8:1932. doi:10.3389/fimmu.2017.01932. [DOI] [PMC free article] [PubMed]
- 128.Li X, Kong D, Chen H, Liu S, Hu H, Wu T et al. miR-155 acts as an anti-inflammatory factor in atherosclerosis-associated foam cell formation by repressing calcium-regulated heat stable protein 1. Sci Rep 2016;6:21789. doi:10.1038/srep21789. [DOI] [PMC free article] [PubMed]
- 129.Huffaker TB, Hu R, Runtsch MC, Bake E, Chen X, Zhao J, et al. Epistasis between microRNAs 155 and 146a during T cell-mediated antitumor immunity. Cell Rep. 2012;2(6):1697–1709. doi: 10.1016/j.celrep.2012.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Ying W, Riopel M, Bandyopadhyay G, Dong Y, Birmingham A, Seo JB et al. Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell. 2017;171(2):372–84.e12. doi:10.1016/j.cell.2017.08.035. [DOI] [PubMed]
- 131.Lin X, Qin Y, Jia J, Lin T, Lin X, Chen L, et al. MiR-155 enhances insulin sensitivity by coordinated regulation of multiple genes in mice. PLoS Genet. 2016;12(10):e1006308. doi: 10.1371/journal.pgen.1006308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ibanez L, Del Rio L, Diaz M, Sebastiani G, Pozo OJ, Lopez-Bermejo A, et al. Normalizing ovulation rate by preferential reduction of hepato-visceral fat in adolescent girls with polycystic ovary syndrome. J Adolesc Health. 2017;61(4):446–453. doi: 10.1016/j.jadohealth.2017.04.010. [DOI] [PubMed] [Google Scholar]
- 133.Coleman CB, Lightell DJ, Jr, Moss SC, Bates M, Parrino PE, Woods TC. Elevation of miR-221 and -222 in the internal mammary arteries of diabetic subjects and normalization with metformin. Mol Cell Endocrinol. 2013;374(1–2):125–129. doi: 10.1016/j.mce.2013.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wehr E, Moller R, Horejsi R, Giuliani A, Kopera D, Schweighofer N, et al. Subcutaneous adipose tissue topography and metabolic disturbances in polycystic ovary syndrome. Wien Klin Wochenschr. 2009;121(7–8):262–269. doi: 10.1007/s00508-009-1162-2. [DOI] [PubMed] [Google Scholar]
- 135.Azziz R, Carmina E, Dewailly D, Diamanti-Kandarakis E, Escobar-Morreale HF, Futterweit W et al. Positions statement: criteria for defining polycystic ovary syndrome as a predominantly hyperandrogenic syndrome: an Androgen Excess Society guideline. J Clin Endocrinol Metab. 2006;91(11):4237–4245. doi:10.1210/jc.2006-0178. [DOI] [PubMed]
- 136.Jonard S, Dewailly D. The follicular excess in polycystic ovaries, due to intra-ovarian hyperandrogenism, may be the main culprit for the follicular arrest. Hum Reprod Update. 2004;10(2):107–117. doi: 10.1093/humupd/dmh010. [DOI] [PubMed] [Google Scholar]
- 137.Zhang G, Garmey JC, Veldhuis JD. Interactive stimulation by luteinizing hormone and insulin of the steroidogenic acute regulatory (StAR) protein and 17alpha-hydroxylase/17,20-lyase (CYP17) genes in porcine theca cells. Endocrinology. 2000;141(8):2735–2742. doi: 10.1210/endo.141.8.7595. [DOI] [PubMed] [Google Scholar]
- 138.Burghen GA, Givens JR, Kitabchi AE. Correlation of hyperandrogenism with hyperinsulinism in polycystic ovarian disease. J Clin Endocrinol Metab. 1980;50(1):113–116. doi: 10.1210/jcem-50-1-113. [DOI] [PubMed] [Google Scholar]
- 139.Geng Y, He J, Ding Y, Chen X, Zhou Y, Liu S et al. The differential expression of microRNAs between implantation sites and interimplantation sites in early pregnancy in mice and their potential functions. Reprod Sci (Thousand Oaks, Calif). 2014;21(10):1296–1306. doi:10.1177/1933719114525273. [DOI] [PubMed]
- 140.Chang EM, Han JE, Seok HH, Lee DR, Yoon TK, Lee WS. Insulin resistance does not affect early embryo development but lowers implantation rate in in vitro maturation-in vitro fertilization-embryo transfer cycle. Clin Endocrinol. 2013;79(1):93–99. doi: 10.1111/cen.12099. [DOI] [PubMed] [Google Scholar]
- 141.Fornes R, Ormazabal P, Rosas C, Gabler F, Vantman D, Romero C et al. Changes in the expression of insulin signaling pathway molecules in endometria from polycystic ovary syndrome women with or without hyperinsulinemia. Mol Med (Cambridge, Mass). 2010;16(3–4):129–136. doi:10.2119/molmed.2009.00118. [DOI] [PMC free article] [PubMed]
- 142.Qi J, Wang W, Zhu Q, He Y, Lu Y, Wang Y, et al. Local cortisol elevation contributes to endometrial insulin resistance in polycystic ovary syndrome. J Clin Endocrinol Metab. 2018;103(7):2457–2467. doi: 10.1210/jc.2017-02459. [DOI] [PubMed] [Google Scholar]
- 143.Altmäe S, Koel M, Võsa U, Adler P, Suhorutšenko M, Laisk-Podar T, et al. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers. Sci Rep. 2017;7(1):10077. doi: 10.1038/s41598-017-10098-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zhao Z, Zhao Q, Warrick J, Lockwood CM, Woodworth A, Moley KH, et al. Circulating microRNA miR-323-3p as a biomarker of ectopic pregnancy. Clin Chem. 2012;58(5):896–905. doi: 10.1373/clinchem.2011.179283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Hu LL, Qian K, Li HX, Sun YP, Zhu GJ. MicroRNA expression and its role in the cell cycle regulation in decidualized endometrial stromal cells in vitro. Zhonghua fu chan ke za zhi. 2012;47(2):129–133. [PubMed] [Google Scholar]
- 146.Homer H, Rice GE, Salomon C. Review: Embryo- and endometrium-derived exosomes and their potential role in assisted reproductive treatments-liquid biopsies for endometrial receptivity. Placenta. 2017;54:89–94. doi: 10.1016/j.placenta.2016.12.011. [DOI] [PubMed] [Google Scholar]
- 147.Li C, Chen L, Zhao Y, Chen S, Fu L, Jiang Y, et al. Altered expression of miRNAs in the uterus from a letrozole-induced rat PCOS model. Gene. 2017;598:20–26. doi: 10.1016/j.gene.2016.10.033. [DOI] [PubMed] [Google Scholar]
- 148.Green CJ, Fraser ST, Day ML. Insulin-like growth factor 1 increases apical fibronectin in blastocysts to increase blastocyst attachment to endometrial epithelial cells in vitro. Hum Reprod (Oxford, England) 2015;30(2):284–298. doi: 10.1093/humrep/deu309. [DOI] [PubMed] [Google Scholar]
- 149.Fazleabas AT, Kim JJ, Srinivasan S, Donnelly KM, Brudney A, Jaffe RC. Implantation in the baboon: endometrial responses. Semin Reprod Endocrinol. 1999;17(3):257–265. doi: 10.1055/s-2007-1016233. [DOI] [PubMed] [Google Scholar]
- 150.Franks S, Mason H, White D, Willis D. Etiology of anovulation in polycystic ovary syndrome. Steroids. 1998;63(5–6):306–307. doi: 10.1016/s0039-128x(98)00035-x. [DOI] [PubMed] [Google Scholar]
- 151.Willis D, Mason H, Gilling-Smith C, Franks S. Modulation by insulin of follicle-stimulating hormone and luteinizing hormone actions in human granulosa cells of normal and polycystic ovaries. J Clin Endocrinol Metab. 1996;81(1):302–309. doi: 10.1210/jcem.81.1.8550768. [DOI] [PubMed] [Google Scholar]
- 152.Bhushan S, Palta P, Bansal N, Sharma V, Manik RS. Effect of insulin on the proliferation of and progesterone production by buffalo granulosa cells in vitro. Vet Rec. 2005;157(23):746–747. doi: 10.1136/vr.157.23.746. [DOI] [PubMed] [Google Scholar]
- 153.Tilly JL. Apoptosis and ovarian function. Rev Reprod. 1996;1(3):162–172. doi: 10.1530/ror.0.0010162. [DOI] [PubMed] [Google Scholar]
- 154.Morita Y, Tilly JL. Oocyte apoptosis: like sand through an hourglass. Dev Biol. 1999;213(1):1–17. doi: 10.1006/dbio.1999.9344. [DOI] [PubMed] [Google Scholar]
- 155.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(6):1219–1227. doi: 10.1007/s00404-017-4523-5. [DOI] [PubMed] [Google Scholar]
- 156.Shalev E, Goldman S, Ben-Shlomo I. The balance between MMP-9 and MMP-2 and their tissue inhibitor (TIMP)-1 in luteinized granulosa cells: comparison between women with PCOS and normal ovulatory women. Mol Hum Reprod. 2001;7(4):325–331. doi: 10.1093/molehr/7.4.325. [DOI] [PubMed] [Google Scholar]
- 157.Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum Reprod Update. 2008;14(2):159–177. doi: 10.1093/humupd/dmm040. [DOI] [PubMed] [Google Scholar]
- 158.Hu MH, Zheng SX, Yin H, Zhu XY, Lu FT, Tong XH et al. Identification of microRNAs that regulate the MAPK pathway in human cumulus cells from PCOS women with insulin resistance. Reprod Sci (Thousand Oaks, Calif). 2020;27(3):833–844. doi:10.1007/s43032-019-00086-5. [DOI] [PubMed]
- 159.Stubbs SA, Webber LJ, Stark J, Rice S, Margara R, Lavery S, et al. Role of insulin-like growth factors in initiation of follicle growth in normal and polycystic human ovaries. J Clin Endocrinol Metab. 2013;98(8):3298–3305. doi: 10.1210/jc.2013-1378. [DOI] [PubMed] [Google Scholar]
- 160.Zhou J, Kumar TR, Matzuk MM, Bondy C. Insulin-like growth factor I regulates gonadotropin responsiveness in the murine ovary. Mol Endocrinol (Baltimore, Md) 1997;11(13):1924–1933. doi: 10.1210/mend.11.13.0032. [DOI] [PubMed] [Google Scholar]
- 161.Desai NA, Patel SS. Increased insulin-like growth factor-1 in relation to cardiovascular function in polycystic ovary syndrome: friend or foe? Gynecol Endocrinol. 2015;31(10):801–807. doi: 10.3109/09513590.2015.1075497. [DOI] [PubMed] [Google Scholar]
- 162.Hasegawa T, Kamada Y, Hosoya T, Fujita S, Nishiyama Y, Iwata N, et al. A regulatory role of androgen in ovarian steroidogenesis by rat granulosa cells. J Steroid Biochem Mol Biol. 2017;172:160–165. doi: 10.1016/j.jsbmb.2017.07.002. [DOI] [PubMed] [Google Scholar]
- 163.Feng R, Sang Q, Zhu Y, Fu W, Liu M, Xu Y, et al. MiRNA-320 in the human follicular fluid is associated with embryo quality in vivo and affects mouse embryonic development in vitro. Sci Rep. 2015;5:8689. doi: 10.1038/srep08689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bhardwaj R, Ansari MM, Pandey S, Parmar MS, Chandra V, Kumar GS, et al. GREM1, EGFR, and HAS2; the oocyte competence markers for improved buffalo embryo production in vitro. Theriogenology. 2016;86(8):2004–2011. doi: 10.1016/j.theriogenology.2016.06.019. [DOI] [PubMed] [Google Scholar]
- 165.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(2):211–221. doi: 10.1007/s10815-018-1335-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Onalan G, Selam B, Baran Y, Cincik M, Onalan R, Gunduz U et al. Serum and follicular fluid levels of soluble Fas, soluble Fas ligand and apoptosis of luteinized granulosa cells in PCOS patients undergoing IVF. Hum Reprod (Oxford, England). 2005;20(9):2391–2395. doi:10.1093/humrep/dei068. [DOI] [PubMed]
- 167.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(3):881–887. doi: 10.1210/jc.2007-1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Erickson GF, Magoffin DA, Garzo VG, Cheung AP, Chang RJ. Granulosa cells of polycystic ovaries: are they normal or abnormal? Hum Reprod (Oxford, England) 1992;7(3):293–299. doi: 10.1093/oxfordjournals.humrep.a137638.. [DOI] [PubMed] [Google Scholar]
- 169.Eppig JJ. Oocyte control of ovarian follicular development and function in mammals. Reprod (Cambridge, England) 2001;122(6):829–838. doi: 10.1530/rep.0.1220829. [DOI] [PubMed] [Google Scholar]
- 170.Hawkins SM, Matzuk MM. Oocyte-somatic cell communication and microRNA function in the ovary. Ann Endocrinol. 2010;71(3):144–148. doi: 10.1016/j.ando.2010.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.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(5):1330–7.e3. doi:10.1016/j.fertnstert.2016.01.020. [DOI] [PubMed]
- 172.Zheng Q, Li Y, Zhang D, Cui X, Dai K, Yang Y, et al. ANP promotes proliferation and inhibits apoptosis of ovarian granulosa cells by NPRA/PGRMC1/EGFR complex and improves ovary functions of PCOS rats. Cell Death Dis. 2017;8(10):e3145. doi: 10.1038/cddis.2017.494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ni XR, Sun ZJ, Hu GH, Wang RH. High concentration of insulin promotes apoptosis of primary cultured rat ovarian granulosa cells via its increase in extracellular HMGB1. Reprod Sci (Thousand Oaks, Calif) 2015;22(3):271–277. doi: 10.1177/1933719114549852. [DOI] [PubMed] [Google Scholar]
- 174.Gao H, Gong N, Ma Z, Miao X, Chen J, Cao Y, et al. LncRNA ZEB2-AS1 promotes pancreatic cancer cell growth and invasion through regulating the miR-204/HMGB1 axis. Int J Biol Macromol. 2018;116:545–551. doi: 10.1016/j.ijbiomac.2018.05.044. [DOI] [PubMed] [Google Scholar]
- 175.Wissing ML, Kristensen SG, Andersen CY, Mikkelsen AL, Host T, Borup R, et al. Identification of new ovulation-related genes in humans by comparing the transcriptome of granulosa cells before and after ovulation triggering in the same controlled ovarian stimulation cycle. Hum Reprod (Oxford, England) 2014;29(5):997–1010. doi: 10.1093/humrep/deu008.. [DOI] [PubMed] [Google Scholar]
- 176.Tosca L, Rame C, Chabrolle C, Tesseraud S, Dupont J. Metformin decreases IGF1-induced cell proliferation and protein synthesis through AMP-activated protein kinase in cultured bovine granulosa cells. Reproduction (Cambridge, England) 2010;139(2):409–418. doi: 10.1530/rep-09-0351. [DOI] [PubMed] [Google Scholar]
- 177.Mani AM, Fenwick MA, Cheng Z, Sharma MK, Singh D, Wathes DC. IGF1 induces up-regulation of steroidogenic and apoptotic regulatory genes via activation of phosphatidylinositol-dependent kinase/AKT in bovine granulosa cells. Reproduction (Cambridge, England) 2010;139(1):139–151. doi:10.1530/rep-09-0050 [DOI] [PubMed]
- 178.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(5):1209–1217. doi: 10.1095/biolreprod62.5.1209. [DOI] [PubMed] [Google Scholar]
- 179.Thierry van Dessel HJ, Lee PD, Faessen G, Fauser BC, Giudice LC. Elevated serum levels of free insulin-like growth factor I in polycystic ovary syndrome. J Clin Endocrinol Metab. 1999;84(9):3030–3035. doi: 10.1210/jcem.84.9.5941. [DOI] [PubMed] [Google Scholar]
- 180.Velazquez MA, Hermann D, Kues WA, Niemann H. Increased apoptosis in bovine blastocysts exposed to high levels of IGF1 is not associated with downregulation of the IGF1 receptor. Reproduction (Cambridge, England) 2011;141(1):91–103. doi: 10.1530/rep-10-0336. [DOI] [PubMed] [Google Scholar]
- 181.Luo L, Wang Q, Chen M, Yuan G, Wang Z, Zhou C. IGF-1 and IGFBP-1 in peripheral blood and decidua of early miscarriages with euploid embryos: comparison between women with and without PCOS. Gynecol Endocrinol. 2016;32(7):538–542. doi: 10.3109/09513590.2016.1138459. [DOI] [PubMed] [Google Scholar]
- 182.Health and fertility in World Health Organization group 2 anovulatory women. Hum Reprod Update. 2012;18(5):586–99. 10.1093/humupd/dms019. [DOI] [PubMed]
- 183.Azziz R, Carmina E, Dewailly D, Diamanti-Kandarakis E, Escobar-Morreale HF, Futterweit W, et al. The Androgen Excess and PCOS Society criteria for the polycystic ovary syndrome: the complete task force report. Fertil Steril. 2009;91(2):456–488. doi: 10.1016/j.fertnstert.2008.06.035. [DOI] [PubMed] [Google Scholar]
- 184.Kuwabara Y, Ono K, Horie T, Nishi H, Nagao K, Kinoshita M, et al. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ Cardiovasc Genet. 2011;4(4):446–454. doi: 10.1161/circgenetics.110.958975. [DOI] [PubMed] [Google Scholar]
- 185.Abbott DH, Bacha F. Ontogeny of polycystic ovary syndrome and insulin resistance in utero and early childhood. Fertil Steril. 2013;100(1):2–11. doi: 10.1016/j.fertnstert.2013.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Brettenthaler N, De Geyter C, Huber PR, Keller U. Effect of the insulin sensitizer pioglitazone on insulin resistance, hyperandrogenism, and ovulatory dysfunction in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2004;89(8):3835–3840. doi: 10.1210/jc.2003-031737. [DOI] [PubMed] [Google Scholar]
- 187.Selimoglu H, Duran C, Kiyici S, Ersoy C, Guclu M, Ozkaya G, et al. The effect of vitamin D replacement therapy on insulin resistance and androgen levels in women with polycystic ovary syndrome. J Endocrinol Investig. 2010;33(4):234–238. doi: 10.3275/6560.10.1007/bf03345785. [DOI] [PubMed] [Google Scholar]
- 188.Velthut-Meikas A, Simm J, Tuuri T, Tapanainen JS, Metsis M, Salumets A. Research resource: small RNA-seq of human granulosa cells reveals miRNAs in FSHR and aromatase genes. Mol Endocrinol (Baltimore, Md) 2013;27(7):1128–1141. doi: 10.1210/me.2013-1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Ding CF, Chen WQ, Zhu YT, Bo YL, Hu HM, Zheng RH. Circulating microRNAs in patients with polycystic ovary syndrome. Human Fertility (Cambridge, England) 2015;18(1):22–29. doi: 10.3109/14647273.2014.956811. [DOI] [PubMed] [Google Scholar]
- 190.Berezikov E. Evolution of microRNA diversity and regulation in animals. Nat Rev Genet. 2011;12(12):846–860. doi: 10.1038/nrg3079. [DOI] [PubMed] [Google Scholar]
- 191.Sun Q, Zhang Y, Yang G, Chen X, Zhang Y, Cao G, et al. Transforming growth factor-beta-regulated miR-24 promotes skeletal muscle differentiation. Nucleic Acids Res. 2008;36(8):2690–2699. doi: 10.1093/nar/gkn032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Sun T, Wang Q, Balk S, Brown M, Lee GS, Kantoff P. The role of microRNA-221 and microRNA-222 in androgen-independent prostate cancer cell lines. Cancer Res. 2009;69(8):3356–3363. doi: 10.1158/0008-5472.Can-08-4112. [DOI] [PMC free article] [PubMed] [Google Scholar]