Skip to main content

Alteration of the N6-methyladenosine methylation landscape in a mouse model of polycystic ovary syndrome

Abstract

Objective

To explore the N6-methyladenosine (m6A) methylation abnormality of mRNAs and its potential roles in the mouse model of polycystic ovary syndrome (PCOS).

Methods

The mouse model of PCOS were induced by injecting dehydroepiandrosterone (DHEA), and confirmed by observing the morphological structures of ovarian follicles. Subsequently, m6A-tagged mRNAs were identified via m6A epitranscriptomic microarray and its potential functional pathways were predicted in KEGG database. The expression and modification levels of key mRNAs in the most enriched pathway were evaluated and compared using western blot and methylated RNA immunoprecipitation-quantitative PCR (MeRIP-qPCR).

Results

Compared with the control group, 415 hypermethylated and downregulated mRNAs, 8 hypomethylated and upregulated mRNAs, and 14 hypermethylated and upregulated mRNAs were identified in the PCOS group (Fold change ≥ 1.5). Those mRNAs were mainly involved in insulin signaling pathway, type II diabetes mellitus, Fc epsilon RI signaling pathway, inositol phosphate metabolism, and GnRH secretion. In insulin signaling pathway, the expression levels of phosphorylated protein kinase B (p-AKT) were decreased, whereas that of upstream phosphorylated phosphatidylinositol 3-kinase (p-PI3K) were increased in PCOS group. Moreover, skeletal muscle and kidney-enriched inositol polyphosphate 5-phosphatease (SKIP), one of PIP3 phosphatases, was verified to be overexpressed, and Skip mRNAs were hypermethylated in PCOS group.

Conclusion

The altered m6A modification of mRNAs might play a critical role in PCOS process. The PI3K/AKT pathway is inhibited in the mouse model of PCOS. Whether it is caused by the m6A modification of Skip mRNAs is worthy of further exploration.

Introduction

Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic dysfunction condition in the women of reproductive age, resulting in irregular menstruation, hyperandrogenism, infertility, and insulin resistance(IR) [1]. Patients with PCOS are at higher risk of diabetes [2], cardiovascular disease [3], and endometrial cance [4]. There is no specific therapy for PCOS, only symptomatic treatment such as lifestyle management, taking combination oral contraceptives to regulate menstrual cycles and ameliorate hyperandrogenism, using insulin sensitizers to alleviate IR, taking clomiphene or letrozole to promote ovulation in the case of anovulatory infertility [5,6,7].

The etiology of PCOS is not yet entirely understood. It is widely assumed that PCOS is caused by a combination of environmental and genetic factors. PCOS is associated with hypothalamic-pituitary-ovarian axis (HPOA) neuroendocrine abnormalities, hyperandrogenemia, IR, chronic inflammation, and circadian rhythm disorder [5, 8]. At the same time, PCOS appears to be a highly genetic and polygenic disease. Despite the fact that candidate gene association studies and genome-wide association studies have identified several susceptibility genes and single-nucleotide polymorphism loci significantly associated with PCOS, such as follicle stimulating hormone receptor gene (FSHR, rs2268361, rs2349415), insulin receptor (INSR, rs2059807), it is still difficult to explain the complexity of PCOS etiology and clinical manifestations [9,10,11]. Recently, epigenetic factors have received a lot of attention in the pathogenesis of PCOS. As far as we know,women with PCOS have different epigenetic regulation, including DNA methylation, histone acetylation, and changes in non coding RNA content [12, 13]. The changes of DNA methylation in peripheral and umbilical cord blood, ovary and adipose tissue of PCOS patients indicate that this epigenetic modification is related to the pathogenesis of the disease [14]. Perhaps, these defects in DNA methylation promote the disorder of genes involved in inflammation, hormone synthesis and signal transduction, as well as glucose and lipid metabolism [15,16,17,18,19]. And the research on the role of DNA methylation in the pathogenesis of PCOS has just begun.

N6-methyladenosine (m6A) is the most prevalent internal modification of mRNAs, which is reversible and dynamically regulated by methyltransferase complexes (writers) and demethylases (erasers), and recognized by m6A binding proteins (readers) [20]. m6A modification regulates post-transcriptional expression of m6A-tagged genes by participating in RNA metabolism such as pre-mRNA splicing, mRNA translation, nuclear export, mRNA decay, and non-coding RNA biogenesis [21]. Recently, studies have revealed the role of m6A modifications and their protein machines in oogenesis and in female reproductive tumors, as well as in other female reproductive diseases [22]. It has been demonstrated that the m6A modification level was increased, and several m6A modulators were dysfunction in PCOS patients [23]. In oogenesis, lack of YTHDF2 leads to failure of m6A modified mRNA degradation, which affecting the oocyte quality [24, 25]. The m6A proteins are also involved in ovulation, and the m6A writers play an important role in oogenesis, but whether they can be used as therapeutic targets for abnormal ovulation remains to be further investigated. Nevertheless, little attention has been paid to the molecular mechanisms of m6A modification in PCOS.

In this study, we aim to investigate the altered m6A modification landscape of mRNAs in the ovaries of PCOS mice using the epitranscriptomic microarray, and to preliminarily explore the potential signal pathways involved in the PCOS process though KEGG analysis.

Materials and methods

The study was approved by the Institutional Review Board (No.2021-S087), and animal experiments were in accordance with the Guide for the Care and Use of Laboratory Animal by International Committees of The Third Xiangya Hospital of Central South University.

Establishment of a mouse model of PCOS

Female C57BL/6J mice aged 7 weeks were acquired from SJA Laboratory Animal Co. Ltd (Hunan China), and adaptively fed for 1 week on 12 h light/ 12 h dark cycle at room temperature (24 ± 3 °C) with a humidity of 45 ± 2%. During the feeding period, vaginal smears were performed daily at 8:00 a.m. to observe the estrous cycle of mice. Ten mice with normal estrous cycle were selected and randomly divided in two groups: the control and PCOS group. In the PCOS group, 6 mg/(100 g·d) dehydroepiandrosterone (DHEA) and 0.2 ml injectable soybean oil were injected subcutaneously into mice daily for 20 consecutive days. Similarly, mice in the control group were injected with 0.2 ml injectable soybean oil daily for 20 days.

Subsequently, five mice with continuous keratosis of vaginal epithelial cells in PCOS group and five mice in control group were randomly sacrificed on the first day. The two ovaries randomly selected mice from each group were embedded in paraffin and sliced for hematoxylin and eosin (HE) staining to observe the morphological changes and confirm the induction of PCOS. And the three remaining mice in each group, the left ovaries were preserved at -80 °C for following N6-methyladenosine detection, the right ovaries were preserved at -80 °C for following western blot.

RNA extraction and quality control

To isolate total RNA, ovary tissues were cut into small pieces and homogenized in TRIzol reagent before being quantified using a NanoDrop ND-1000 (Thermo Fisher Scientific, Waltham, MA, USA). Supplementary Table S1 presents the quantification and quality of RNA.

m6A immunoprecipitation

Total RNA (1-3ug) mixed with m6A spike-in control was immunoprecipitated with anti-m6A rabbit polyclonal antibody (Synaptic Systems, Göttingen, Germany) at 4 °C for 2 h. Dynabeads™ M-280 Sheep Anti-Rabbit IgG suspension (20 μL per sample) (Ivitrogen) was blocked with 0.5% bovine serum albumin (BSA) at 4 °C for 2 h, washed three times with IP buffer (300 μL) for 5 min, and resuspended in the prepared RNA-antibody mixture. The RNA was bound to the m6A-antibody beads for 2 h at 4 °C, then the beads were washed with IP buffer (500 μL, three times), followed by Wash buffer (500 μL, twice). In this case, the adsorbed RNA was eluted with Elution buffer (200 μL) at 50 °C for 1 h. The immunoprecipitated (IP) RNA and supernatant (Sup) RNA were extracted by acid phenol–chloroform and ethanol precipitation.

Labeling and hybridization

The IP RNAs and Sup RNAs were mixed with an equal amount of calibration spike-in control RNA, amplified separately and labeled with Cy3 (for Sup) and Cy5 (for IP) using Arraystar Super RNA Labeling Kit (Arraystar). The synthesized cRNAs was purified by the RNeasy Mini Kit (QIAGEN), and the concentration and specific activity of cRNAs were detected by the NanoDrop ND-1000 (Thermo Fisher Scientific) (Supplementary Table S2). 2.5 μg of Cy3 and Cy5-labeled cRNAs were mixed, added with 5 μL 10 × Blocking Agent and 1 μL of 25 × Fragmentation Buffer. It was heated to 60℃ for 30 min, and then mixed with 25 μL 2 × Hybridization buffer. 50 μl of hybridization solution was injected into the gasket slide and assembled on the m6A-mRNA epitranscriptome microarray slide. The slides were incubated at 65 °C for 17 h in an Agilent Hybridization Oven (Agilent, CA, USA). The hybridized arrays were washed, fixed, and scanned with an Agilent scanner G2505C (Agilent).

Epitranscriptomic microarray data analysis

To analyze acquired array images, Agilent Feature Extraction software (version 11.0.1.1) was used. The raw IP and Sup intensities were normalized to the log2-scaled Spike-in RNA intensity average. Following Spike-in normalization, probe signals with Present (P) or Marginal (M) QC flags were retained in at least three of six samples for further m6A methylation, quantity, and expression level analyses. The m6A methylation level was calculated for the percentage of modification based on the IP and Sup normalized intensities, and the m6A quantity was calculated for the m6A methylation amount. The expression level was calculated by adding the IP and Sup normalized intensities, and an additional quantile normalization method from the limma package was used to normalize the RNA expression level between arrays before flagging probes.

Methylated RNA immunoprecipitation-quantitative PCR (MeRIP-qPCR)

The m6A epitranscriptomic microarray data was validated using IP (n = 3, each group). MeRIP-qPCR was then used to quantify the RNA enrichment via 2−ΔΔct analysis. Supplementary Table S3 describes the primer used. Furthermore, the mRNA m6A sites were predicted using the sequence-based RNA adenosine methylation site predictor (SRAMP) program (https://www.cuilab.cn/sramp) [26].

Western blot

The protein concentration of right ovary tissue was determined using the Bradford method after protein extraction (M&C Gene Technology Ltd.). SDS–polyacrylamide gel electrophoresis (SDS-PAGE) was used to separate the protein samples, which were then transferred to a polyvinylidene difluoride membrane. Antibodies of interest were used to probe the membranes. The antibodies used were as follows: phosphatidylinositol 3-kinase (PI3K) (1:1000; ABclonal), phosphorylated PI3K (p-PI3K) (1:1000; ABclonal), protein kinase B (AKT) (1:1000; Proteintech), phosphorylated AKT (ser473) (p-AKT) (1:1000; Proteintech), skeletal muscle and kidney-enriched inositol polyphosphate 5-phosphatase (SKIP) (1:1000; Proteintech), and β-actin (1:1000; Abcam).

Statistical analysis

Filtering with the fold change ≥ 1.5 and statistical significance thresholds (P < 0.05) revealed differentially m6A-methylated or differentially expressed RNAs between two comparison groups. Hierarchical clustering was carried out using the R software (version 4.02). To perform GO analysis, the topGO package in the R environment for statistical computing and graphics was used, and the Fisher's exact test was used to perform pathway analysis. The western blot data were presented as mean ± SD and compared between groups using the Student's t test or the Mann–Whitney U test. P < 0.05 was regarded as significant. SPSS version 21.0 was used for all statistical analysis (SPSS 21.0, Inc.,Chicago, IL, USA).

Results

Mouse models of PCOS

In the control group, different stages of follicular development were seen in ovaries, and the ovarian morphology was normal without large cysts (Fig. 1A); While the ovaries were swollen with multiple cystic follicles in the PCOS group, and there were cyst-like expanded follicles on the surface of ovaries (Fig. 1B). These indicated that the mouse model of PCOS was successfully established.

Fig. 1
figure 1

The hematoxylin and eosin (HE) staining of ovaries of mouse model in two groups. A the control group; B the PCOS group. *: developing follicles; #: corpus luteum; + : cystic follicles

Identification of differentially expressed or methylated genes

The microarray results showed that 307 mRNAs were significantly differentially expressed (Fold change ≥ 1.5, P value < 0.05) between the PCOS and control groups. Among them, 226 and 81 mRNAs were upregulated and downregulated in the PCOS group, respectively (Fig. 2A and B). Ten most significantly differentially regulated mRNAs are listed respectively in Table 1.

Fig. 2
figure 2

The volcano plot and heatmap plot of differentially expressed or methylated mRNAs. A volcano plot of differentially expressed mRNAs. B heatmap plot of differentially expressed mRNAs. C volcano plot of differentially methylated mRNAs. D heatmap plot of differentially methylated mRNAs

Table 1 Significantly differentially expressed mRNAs

Comparing with the m6A levels in the control groups, 143 mRNAs had significantly differential modification levels (Fold change ≥ 1.5, P value < 0.05) in the PCOS group. Surprisingly, of these 143 mRNAs, 134 mRNAs were found to have higher m6A methylation levels and only 9 mRNAs had lower levels of m6A methylation (Fig. 2C and D). Ten most significantly differentially m6A-methylated mRNAs are listed in Table 2.

Table 2 Significantly differentially methylated mRNAs

Moreover, we found several “writers” and “readers” of m6A modification were up-regulated in PCOS group, including YTH N6-methyladenosine RNA binding protein 3 (Ythdf3) (Foldchange = 1.35, P < 0.05), heterogeneous nuclear ribonucleoprotein A2/B1 (Hnrnpa2b1) (Foldchange = 1.615713434, P < 0.05).

Analysis of differentially expressed with differentially methylated genes

When we integrated the mRNAs methylation and expression data, 8 hypomethylated and upregulated mRNAs, 415 hypermethylated and downregulated mRNAs, 14 hypermethylated and upregulated mRNAs, and 0 hypomethylated and downregulated mRNAs (Fold change ≥ 1.5) were identified (Fig. 3A). Then, the functions of these 437 mRNAs were analyzed through GO and KEGG pathway analyses. Among the enriched GO terms, “cellular process” in biological process (BP), “cellular anatomical entity” in cellular components (CC), “binding” in molecular function (MF) earned the highest enrichment score (Fig. 3B). As for the KEGG analysis, mRNAs were predicted to participated in 24 pathways and the most enriched ten pathways are shown in Fig. 3C. Insulin signaling pathway, the most enriched pathway, involved ten differentially expressed mRNAs with differentially methylation (Fig. 3D).

Fig. 3
figure 3

Analysis of differentially expressed with differentially methylated genes. A. scatter plot; B. the top ten enriched items obtained from GO analysis; C. the first ten enriched pathways identified in KEGG analysis. D. the most enriched pathway-insulin signaling pathway. mRNAs with different expression and m6A modification levels were marked in orange. The diagram is based on the insulin signaling pathway in the Kyoto encyclopedia of genes and genomes pathway database

Among these, the higher m6A methylation levels of the skeletal muscle and kidney-enriched inositol polyphosphate 5-phosphatase (Skip) mRNA were verified via MeRIP-PCR (P < 0.01) (Fig. 4). Moreover, analysis of Skip mRNA with SRAMP program predicted six potential m6A sites with very high confidence, including five sites on the coding sequence and one sites on the 3’untranslated region (UTR) (Fig. 4, Supplementary Table 4).

Fig. 4
figure 4

The comparison of m6 A modification levels and the predicted m6 A site of Skip mRNAThe left image shows that the m6 A modification levels were higher in the PCOS group than that in the control group (P < 0.01); The right image shows that the predicted m.6 A sites by SRAMP program (https://www.cuilab.cn/sramp)

Suppressed phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (AKT) signaling pathway

We further explored the activity levels of PI3K/AKT signaling pathway by western blotting, which is one part of insulin signaling pathway. It demonstrated that the expression of AKT and p-AKT (ser473) were significantly decreased (P < 0.001), whereas the expression of PI3K and p-PI3K were significantly increased (P < 0.05) in PCOS group than that in control group (Fig. 5). Notably, the SKIP, a phosphatidylinositol 3,4,5-trisphpsphate (PIP3) phosphatase, was identified to be overexpressed in PCOS group (P < 0.001) (Fig. 5).

Fig. 5
figure 5

Suppressed PI3K/AKT signaling pathway and elevated SKIP expression. The top image shows the representative image of western blotting in control and PCOS group; The remaining images shows the comparison of PI3K, p-PI3K, AKT, p-AKT, and SKIP expression levels between two groups. Data were shown as the mean ± SD. All experiment was conducted in triple. *P < 0 .05, **P < 0.01 and ***P < 0.001

Discussion

Polycystic ovary syndrome (PCOS) has numerous adverse effects on women. However, the pathogenesis of PCOS is currently unclear, and there is no specific therapy for PCOS. Given the continuously increasing incidence of PCOS in recent years, it is important to explore the mechanisms of PCOS development and to develop effective treatments for patients with PCOS. PCOS is commonly associated with aberrant DNA methylation, and several genes are epigenetically dysregulated, and are associated with the pathological consequences of PCOS and metabolic comorbidities [27]. However, the methylation status of specific genes and the extent to which genes are dysregulated in terms of methylation patterns are unknown.Due to the reversibility of epigenetic modifications, "druable" regions can be screened to target or correct abnormalities in gene expression, so PCOS methylation promises the development of novel chromatin methylation therapies targeting PCOS [28].Herein, we used epitranscriptomic microarray for the first time to investigate the altered m6A modification of mRNAs and preliminarily explore the potential molecular mechanisms of m6A modification in the mouse model of PCOS induced by hyperandrogenism, and to screen new molecular targets and develop effective targets for treating PCOS or inhibiting its progression.

According to our microarray results, the m6A modification levels of mRNAs were increased, and the m6A ‘readers’ (Ythdf3, Hnrnpa2b1) are overexpressed in PCOS mice ovaries. A similar methylation trend was confirmed using MeRIP sequencing (MeRIP-seq) in luteinized granulosa cells (GCs) of PCOS patients [23]. In contrast to MeRIP-seq, epitranscriptomic microarray can determined the percentage of modified and unmodified RNA of each transcript [29]. As a result, 437 RNAs with differentially expressed and differentially methylated levels were identified. Further bioinformatics analysis revealed that the m6A modification may mainly participate in the insulin signaling pathway and type II diabetes mellitus pathway, which were closely related to IR and secondary hyperinsulinemia [30]. The PI3K/AKT pathway and the mitogen activated protein kinase (MAPK) pathway were two major insulin-related signal transduction pathway, regulating the glucose metabolism, cell proliferation and differentiation, respectively [30, 31]. In mouse cumulus-oocyte complexes, activated PI3K/AKT signaling can increase glucose uptake by mediating the translocation of GLUT4 to the GCs membrane, which provides energy substrate for follicular development [32]. The dysfunction of PI3K/AKT signaling is not only linked to IR, inflammation, and oxidative stress, but it also inhibits proliferation and promotes apoptosis, all of which may contribute to PCOS [33, 34]. Similarly, the inhibition of PI3K/AKT pathway in PCOS mice was confirmed in our study by assessing p-AKT expression levels.

Notably, the activity of upstream and downstream factors in the PI3K/AKT pathway were opposite. In contrast to p-AKT, p-PI3K expression levels were elevated in the PCOS group, which may be related to inositol phosphate metabolism pathway, another enriched pathway. SKIP, a PIP3 5-phosphatase, was localized at endoplasmic reticulum under resting conditions. Insulin stimulation induced its translocation to the plasma membrane, and binding with activated p21-activated protein kinase 1 (PAK1), thereby activating SKIP’s PIP3 phosphatase activity. The rapid and efficient hydrolysis of PIP3, resulting in decreased AKT2 phosphorylation, inhibits membrane ruffle and GLUT4 translocation, thus negatively regulates insulin signaling in skeletal muscle [35, 36]. As expected, SKIP protein was confirmed to be significantly overexpressed in PCOS mice in this study. Furthermore, due to the non-statistically significant difference in mRNA expression levels between two groups, SKIP overexpression appears to be linked to higher m6A modification in PCOS mice. The m6A sites in humans and mice are highly conserved, and mainly enriched in the 3’untranslated region (UTR) and around stop codons [37]. However, Shen Zhang et al. discovered increased m6A peaks in the coding sequence and transcription start regions, but less prominent enrichment near stop codons in PCOS patients' GCs compared to controls [23]. Similarly, the m6A sites of Skip mRNA was also mainly predicted in the coding sequence by SRAMP program in this study [26]. In addition, previous research revealed that the METTL3-induced m6A modification in the coding sequence may alleviate ribosome stalling and thus improve mRNA translation in acute myeloid leukemia [38]. And in Arabidopsis thaliana, increased mRNA expression levels were associated with a modification change around the start codon [39]. Therefore, the altered m6A modification of Skip mRNA may explain its translation upregulation. Therefore, it is possible that the higher m6A modification of Skip mRNA result in the overexpression of SKIP protein, then reversed the activation of p-PI3K on the downstream AKT.

In addition, Fc epsilon RI signaling pathway and GnRH secretion pathway, another two enriched pathways, may indicated that the m6A modification also participant in the etiological mechanism of chronic inflammation and neuroendocrine in PCOS. Increasing number of studies demonstrated that low-grade chronic inflammation can induce IR, obesity, and hyperandrogenemia through related pathways, leading to ovulation disorders in PCOS [40,41,42]. The HPOA plays an important role in the regulation of female reproductive endocrine as an integrated and coordinated neuroendocrine system. Hypothalamic gonadotropin-releasing hormone (GnRH) neurons secrete GnRH, which regulates the secretion of gonadotropins FSH and LH, then regulate the secretion of sex hormones and reproductive function [43]. The GnRH neuronal firing activity was identified to be increased in PCOS mice induced by prenatal androgenization compared to normal mice, which may be closely linked to PCOS development [44, 45].

Inevitably, there were several limitations of this study. First, several acknowledged inhibiting factors for PI3K/AKT pathway were not investigated, such as Phosphatase and tensin homolog (PTEN) and c-Jun N-terminal kinase (JNK). Then, we did not further validate our hypothesis that the hypermethylation of SKIP mRNAs enhances its expression, then inhibits the PI3K/AKT signaling pathway.

In conclusion, our study demonstrates that the altered m6A modification of mRNAs might play a critical role in PCOS process. And we emphasized the changes in the activity of upstream and downstream factors in the PI3K/AKT signaling pathway. Moreover, the role of m6A modification of Skip mRNA in the pathogenesis of PCOS warrants further studies.

Availability of data and materials

The data and material presented in this manuscript is available from the corresponding author on reasonable request.

References

  1. Kostroun KE, Goldrick K, Mondshine JN, Robinson RD, Mankus E, Reddy S, et al. Impact of updated international diagnostic criteria for the diagnosis of polycystic ovary syndrome. F S Rep. 2023;4:173–8.

    PubMed  Google Scholar 

  2. Livadas S, Paparodis R, Anagnostis P, Gambineri A, Bjekic-Macut J, Petrovic T, et al. Assessment of type 2 diabetes risk in young women with polycystic ovary syndrome. Diagnostics (Basel). 2023;13:2067.

    Article  CAS  PubMed  Google Scholar 

  3. Lo A, Lo C, Oliver-Williams C. Cardiovascular disease risk in women with hyperandrogenism, oligomenorrhea/menstrual irregularity or polycystic ovaries (components of polycystic ovary syndrome): a systematic review and meta-analysis. Eur Heart J Open. 2023;3: d61.

    Article  Google Scholar 

  4. Allen LA, Shrikrishnapalasuriyar N, Rees DA. Long-term health outcomes in young women with polycystic ovary syndrome: A narrative review. Clin Endocrinol (Oxf). 2022;97:187–98.

    Article  PubMed  Google Scholar 

  5. Zaib S, Rana N, Khan I, Waris A, Ahmad U. Analyzing the challenges, consequences, and possible treatments for polycystic ovary syndrome. Mini Rev Med Chem 2023.

  6. Moran LJ, Tassone EC, Boyle J, Brennan L, Harrison CL, Hirschberg AL, et al. Evidence summaries and recommendations from the international evidence-based guideline for the assessment and management of polycystic ovary syndrome: Lifestyle management. Obes Rev. 2020;21: e13046.

    Article  PubMed  Google Scholar 

  7. Sadeghi HM, Adeli I, Calina D, Docea AO, Mousavi T, Daniali M, et al. Polycystic ovary syndrome: a comprehensive review of pathogenesis, management, and drug repurposing. Int J Mol Sci. 2022;23:583.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wang J, Wu D, Guo H, Li M. Hyperandrogenemia and insulin resistance: The chief culprit of polycystic ovary syndrome. Life Sci. 2019;236: 116940.

    Article  CAS  PubMed  Google Scholar 

  9. Wu Y, Yang L, Wu X, Wang L, Qi H, Feng Q, et al. Identification of the hub genes in polycystic ovary syndrome based on disease-associated molecule network. Faseb J. 2023;37: e23056.

    Article  CAS  PubMed  Google Scholar 

  10. Du T, Duan Y, Li K, Zhao X, Ni R, Li Y, et al. Statistical genomic approach identifies association between FSHR polymorphisms and polycystic ovary morphology in women with polycystic ovary syndrome. Biomed Res Int. 2015;2015: 483726.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Cui L, Li G, Zhong W, Bian Y, Su S, Sheng Y, et al. Polycystic ovary syndrome susceptibility single nucleotide polymorphisms in women with a single PCOS clinical feature. Hum Reprod. 2015;30:732–6.

    Article  CAS  PubMed  Google Scholar 

  12. Vazquez-Martinez ER, Gomez-Viais YI, Garcia-Gomez E, Reyes-Mayoral C, Reyes-Munoz E, Camacho-Arroyo I, et al. DNA methylation in the pathogenesis of polycystic ovary syndrome. Reproduction. 2019;158:R27-40.

    Article  CAS  PubMed  Google Scholar 

  13. Miranda AG, Seneda MM, Faustino LR. DNA methylation associated with polycystic ovary syndrome: a systematic review. Arch Gynecol Obstet. 2023.

  14. Concha CF, Sir PT, Recabarren SE. Perez BF [Epigenetics of polycystic ovary syndrome]. Rev Med Chil. 2017;145:907–15.

    Google Scholar 

  15. Szukiewicz D, Trojanowski S, Kociszewska A, Szewczyk G. Modulation of the inflammatory response in polycystic ovary syndrome (PCOS)-searching for epigenetic factors. Int J Mol Sci. 2022;23:14663.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Divoux A, Erdos E, Whytock K, Osborne TF, Smith SR. Transcriptional and DNA methylation signatures of subcutaneous adipose tissue and adipose-derived stem cells in PCOS women. Cells-Basel. 2022;11:848.

    Article  CAS  Google Scholar 

  17. Cao P, Yang W, Wang P, Li X, Nashun B. Characterization of DNA methylation and screening of epigenetic markers in polycystic ovary syndrome. Front Cell Dev Biol. 2021;9: 664843.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Bril F, Ezeh U, Amiri M, Hatoum S, Pace L, Chen YH, et al. Adipose tissue dysfunction in polycystic ovary syndrome. J Clin Endocrinol Metab. 2023;11:179.

    Google Scholar 

  19. Ray RP, Padhi M, Jena S, Patnaik R, Rattan R, Nayak AK. Study of association of global deoxyribonucleic acid methylation in women with polycystic ovary syndrome. J Hum Reprod Sci. 2022;15:233–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Meyer KD, Jaffrey SR. Rethinking m(6)A readers, writers, and erasers. Annu Rev Cell Dev Biol. 2017;33:319–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 2017;18:31–42.

    Article  CAS  PubMed  Google Scholar 

  22. Chen J, Fang Y, Xu Y, Sun H. Role of m6A modification in female infertility and reproductive system diseases. Int J Biol Sci. 2022;18:3592–604.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang S, Deng W, Liu Q, Wang P, Yang W, Ni W. Altered m(6) A modification is involved in up-regulated expression of FOXO3 in luteinized granulosa cells of non-obese polycystic ovary syndrome patients. J Cell Mol Med. 2020;24:11874–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sun X, Lu J, Li H, Huang B. The role of m(6)A on female reproduction and fertility: from gonad development to ovarian aging. Front Cell Dev Biol. 2022;10: 884295.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ivanova I, Much C, Di Giacomo M, Azzi C, Morgan M, Moreira PN, et al. The RNA m(6)A reader YTHDF2 is essential for the post-transcriptional regulation of the maternal transcriptome and oocyte competence. Mol Cell. 2017;67:1059–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhou Y, Zeng P, Li YH, Zhang Z, Cui Q. SRAMP: prediction of mammalian N6-methyladenosine (m6A) sites based on sequence-derived features. Nucleic Acids Res. 2016;44: e91.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Rawat K, Sandhu A, Gautam V, Saha PK, Saha L. Role of genomic DNA methylation in PCOS pathogenesis: a systematic review and meta-analysis involving case-controlled clinical studies. Mol Hum Reprod. 2022;28:gaac024.

    Article  PubMed  Google Scholar 

  28. Smirnov VV, Beeraka NM, Butko DY, Nikolenko VN, Bondarev SA, Achkasov EE, et al. Updates on molecular targets and epigenetic-based therapies for PCOS. Reprod Sci. 2023;30:772–86.

    Article  CAS  PubMed  Google Scholar 

  29. Liu N, Parisien M, Dai Q, Zheng G, He C, Pan T. Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA. RNA. 2013;19:1848–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Beale EG. Insulin signaling and insulin resistance. J Investig Med. 2013;61:11–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Guo S. Insulin signaling, resistance, and the metabolic syndrome: insights from mouse models into disease mechanisms. J Endocrinol. 2014;220:T1-23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Purcell SH, Chi MM, Lanzendorf S, Moley KH. Insulin-stimulated glucose uptake occurs in specialized cells within the cumulus oocyte complex. Endocrinology. 2012;153:2444–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chen C, Jiang X, Ding C, Sun X, Wan L, Wang C. Downregulated lncRNA HOTAIR ameliorates polycystic ovaries syndrome via IGF-1 mediated PI3K/Akt pathway. Gynecol Endocrinol. 2023;39:2227280.

    Article  PubMed  Google Scholar 

  34. Li T, Mo H, Chen W, Li L, Xiao Y, Zhang J, et al. Role of the PI3K-Akt signaling pathway in the pathogenesis of polycystic ovary syndrome. Reprod Sci. 2017;24:646–55.

    Article  CAS  PubMed  Google Scholar 

  35. Ijuin T, Hosooka T, Takenawa T. Phosphatidylinositol 3,4,5-trisphosphate phosphatase SKIP links endoplasmic reticulum stress in skeletal muscle to insulin resistance. Mol Cell Biol. 2016;36:108–18.

    Article  CAS  PubMed  Google Scholar 

  36. Ijuin T, Hatano N, Takenawa T. Glucose-regulated protein 78 (GRP78) binds directly to PIP3 phosphatase SKIP and determines its localization. Genes Cells. 2016;21:457–65.

    Article  CAS  PubMed  Google Scholar 

  37. Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell. 2012;149:1635–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Barbieri I, Tzelepis K, Pandolfini L, Shi J, Millan-Zambrano G, Robson SC, et al. Promoter-bound METTL3 maintains myeloid leukaemia by m(6)A-dependent translation control. Nature. 2017;552:126–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Luo GZ, Macqueen A, Zheng G, Duan H, Dore LC, Lu Z, et al. Unique features of the m6A methylome in Arabidopsis thaliana. Nat Commun. 2014;5:5630.

    Article  CAS  PubMed  Google Scholar 

  40. Popovic M, Sartorius G, Christ-Crain M. Chronic low-grade inflammation in polycystic ovary syndrome: is there a (patho)-physiological role for interleukin-1? Semin Immunopathol. 2019;41:447–59.

    Article  PubMed  Google Scholar 

  41. Rostamtabar M, Esmaeilzadeh S, Tourani M, Rahmani A, Baee M, Shirafkan F, et al. Pathophysiological roles of chronic low-grade inflammation mediators in polycystic ovary syndrome. J Cell Physiol. 2021;236:824–38.

    Article  CAS  PubMed  Google Scholar 

  42. Rudnicka E, Suchta K, Grymowicz M, Calik-Ksepka A, Smolarczyk K, Duszewska AM, et al. Chronic low grade inflammation in pathogenesis of PCOS. Int J Mol Sci. 2021;22:3789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Silva M, Desroziers E, Hessler S, Prescott M, Coyle C, Herbison AE, et al. Activation of arcuate nucleus GABA neurons promotes luteinizing hormone secretion and reproductive dysfunction: Implications for polycystic ovary syndrome. EBioMedicine. 2019;44:582–96.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Moore AM, Prescott M, Marshall CJ, Yip SH, Campbell RE. Enhancement of a robust arcuate GABAergic input to gonadotropin-releasing hormone neurons in a model of polycystic ovarian syndrome. Proc Natl Acad Sci U S A. 2015;112:596–601.

    Article  CAS  PubMed  Google Scholar 

  45. Yan X, Yuan C, Zhao N, Cui Y, Liu J. Prenatal androgen excess enhances stimulation of the GNRH pulse in pubertal female rats. J Endocrinol. 2014;222:73–85.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

Supported by the Young Scientists Fund of the Hunan Provincial Natural Science Foundation of China (Grant No. 2021JJ40958) and the Changsha Natural Science Foundation (Grant No.kk2202425).

Author information

Authors and Affiliations

Authors

Contributions

LZ and WL wrote the manuscript. SZ and BJ conceived the study and edited the manuscript. BJ provided Administrative support. All authors final approval of manuscript.

Corresponding authors

Correspondence to Shujuan Zhu or Bin Jiang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

Additional file 1:

 Supplementary Table S1. the quantification and quality of RNA in ovary tissue of a mouse model of PCOS*. Supplementary Table S2. The specific activity (pmol dyes per μg cRNA) of the labeled RNA. Supplementary Table S3. Primers used in MeRIP-qPCR. Supplementary Table S4. Predicted m6A sites in Skip mRNA by SRAMP program.

Additional file 2:

 Supplementary Figure S1. Characteristics of vaginal smears during various stages of estrus cycle  A. Proestrous, which was characterized by the presence of mostly nucleated and some cornified epithelial cells; B. Estrous, which was characterized by the presence of mostly cornified epithelial cells; C. Metestrus, which was characterized by the presence of cornified epithelial cells and leukocytes; D. Diestrus, which was characterized by the presence of primarily leukocytes.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zou, L., Li, W., Xu, D. et al. Alteration of the N6-methyladenosine methylation landscape in a mouse model of polycystic ovary syndrome. J Ovarian Res 16, 157 (2023). https://doi.org/10.1186/s13048-023-01246-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13048-023-01246-7

Keywords