Abstract

Context

Polycystic ovary syndrome (PCOS) is a chronic disease affecting reproductive function and whole-body metabolism. Although the etiology is unclear, emerging evidence indicates that the epigenetics may be a contributing factor.

Objective

To determine the role of global and genome-wide epigenetic modifications in specific immune cells in PCOS compared with controls and whether these could be related to clinical features of PCOS.

Design

Cross-sectional study.

Participants

Women with (n = 17) or without PCOS (n = 17).

Setting

Recruited from the general community.

Main Outcome Measures

Isolated peripheral blood mononuclear cells were analyzed using multicolor flow cytometry methods to determine global DNA methylation levels in a cell-specific fashion. Transcriptomic and genome-wide DNA methylation analyses were performed on T helper cells using RNA sequencing and reduced representation bisulfite sequencing.

Results

Women with PCOS had lower global DNA methylation in monocytes (P = 0.006) and in T helper (P = 0.004), T cytotoxic (P = 0.004), and B cells (P = 0.03). Specific genome-wide DNA methylation analysis of T helper cells from women with PCOS identified 5581 differentially methylated CpG sites. Functional gene ontology enrichment analysis showed that genes located at the proximity of differentially methylated CpG sites belong to pathways related to reproductive function and immune cell function. However, these genes were not altered at the transcriptomic level.

Conclusions

It was shown that PCOS is associated with global and gene-specific DNA methylation remodeling in a cell type–specific manner. Further investigation is warranted to determine whether epigenetic reprogramming of immune cells is important in determining the different phenotypes of PCOS.

Polycystic ovary syndrome (PCOS) is a metabolic and reproductive disorder with a complex and ill-defined etiology. It is commonly characterized by increased levels of androgens, inflammatory cytokines, insulin resistance, and an increased prevalence of subfertility (13). Familial clustering of PCOS is well documented, providing evidence for a heritable component of the syndrome (4, 5). However, investigations to identify the single-nucleotide polymorphisms that may underpin the genetic basis of PCOS have been inconclusive despite recent advances in the field from genome-wide association studies (69).

Emerging evidence suggests a role for altered epigenetic programming in the etiology of PCOS (1014). Genome-wide analysis of transcriptomic and DNA methylation status have been conducted in adipose tissue and skeletal muscle revealing tissue-specific epigenetic, as well as transcriptomic differences between women with and without PCOS (13, 14). In adipose tissue, DNA methylation analysis revealed aberrant patterns in pathways involved in adipogenesis, inflammation, glucose regulation, and energy metabolism, and these DNA methylation sites corresponded with 30 differentially expressed genes (13). Similarly, in skeletal muscle, transcriptomic analysis found significantly enriched pathways in immune function and disease (14). Finally, both adipose tissue and skeletal muscle showed inflammation as a significantly enriched pathway. This is important because immune cells play a critical role in metabolism and reproductive function, are the main source of inflammatory cytokines, and are known to be epigenetically regulated (1519). These studies provide important insights into the potential contribution of epigenetic changes to the development of PCOS and rationale for further investigation into epigenetic changes in other cell types that may be affected in PCOS, such as immune cells. In both animal PCOS models, and in women with PCOS, altered immune responses and inflammatory markers have been found in various ovarian tissues, suggesting a role in the pathophysiology of the reproductive features of PCOS (2022). Impairment of immune cell function and the infiltration of these cells in insulin-sensitive tissues have been shown to cause metabolic impairment and contribute to the onset of type 2 diabetes mellitus (T2DM) (23, 24). Thus, epigenetic remodeling of immune cells can be associated with impaired immune function and contribute to metabolic dysfunction in PCOS (15, 25, 26).

Only one study has investigated the contribution of global epigenetic changes in immune cells from women with PCOS (27). In that study, the authors measured global DNA methylation (i.e., total, non–site-specific methyl-cytosine abundance) in a mixed population of immune cells known as peripheral blood mononuclear cells (PBMCs) from women with or without PCOS and failed to find any difference (27). There is increasing evidence supporting that epigenetic changes that occur within specific cell subtypes could be masked by analyses of whole blood (28). Therefore, cell type–specific global and genome-wide methylation analyses may reveal a potential role of epigenetics in the immune system, especially in PCOS (2830). In the current study we hypothesized that specific immune cell populations will present a different DNA methylation profile in women with PCOS compared with women without PCOS (25, 28). Using global and genome-wide DNA methylation analyses, we aimed to explore epigenetic remodeling in specific immune cell subtypes, most notably in T helper cells.

Materials and Methods

Study population

Premenopausal women (n = 34) aged between 18 and 45 years with or without PCOS were recruited from the local community (i.e., a nonclinical population). All participants provided informed written consent. Women with PCOS were confirmed by an endocrinologist (Dr. S. Shorakae or Dr. A. Joham) to have features consistent with the Rotterdam diagnostic criteria based on participants’ previous medical records. The Rotterdam criteria were used for confirmation of PCOS with two of the following: (i) oligoovulation or anovulation, (ii) clinical (hirsutism and acne) and/or biochemical hyperandrogenism, and (iii) polycystic ovaries on ultrasound and exclusion of other causes of hyperandrogenism (31, 32). Women without PCOS had no features of PCOS. Exclusion criteria were pregnancy, smoking, T2DM, known cardiovascular disease, asthma, and medications affecting endpoint measures, including hormonal contraceptives, insulin-sensitizing drugs, anti-inflammatories, and antiandrogens. This study was approved by the Victoria University Human Research Ethics Committee (HRE 14-138).

Clinical measures

All clinical measures were collected in the morning after an overnight fast. For women without PCOS, testing was conducted in the early follicular phase of the menstrual cycle (2 to 7 days after beginning menses). All women with PCOS had irregular cycles, and therefore the testing was conducted in early follicular phase when possible.

Lifestyle monitoring

In the 7 days preceding the trial, participants recorded their dietary intake via a 7-day food diary. Food diaries were analyzed by FoodWorks® (Xyris) for the major food groups (grains, fruit, vegetables, protein, and dairy), total energy (macronutrients), and fat ratios. Moderate-to-vigorous physical activity was measured via an ActiGraph (GTX3+) accelerometer. Participants wore the monitor on their right hip during waking hours for 7 days, except for bathing and swimming. Data were downloaded in 1-minute epochs, and nonwear time was defined as at least 20 minutes of zero counts. Daily moderate-to-vigorous physical activity was defined as all wear-time minutes and was calculated based on the Freedson accelerometer age cut-points (33). Self-reporting physical activity was measured via the International Physical Activity Questionnaire.

Anthropometric assessment

Height and weight were measured to calculate body mass index (BMI): [body weight (kg)/ height (m2)]. Waist and hip circumference measurements were recorded (34). Fat mass, abdominal fat mass, and fat free mass were measured by dual-energy x-ray absorptiometry (GE Lunar iDXA) and analyzed by a qualified dual-energy x-ray absorptiometry operator (D.H.).

Fasting blood samples

A sterile polyethylene catheter was inserted into the antecubital vein and baseline blood samples were collected. Plasma and serum were collected in the appropriate blood tubes via centrifugation and stored at −80°C until analysis. Blood for PBMCs isolation was collected in three acid citrate dextrose tubes (BD Biosciences) and left at room temperature until Ficoll gradient separation on the same day as described below (28).

Oral glucose tolerance test

After collecting fasting baseline blood samples, participants then underwent an oral glucose tolerance test (OGTT) via ingestion of a 75-g glucose drink during a 5-minute period with blood samples collected at 30, 60, 90, and 120 minutes (35). Blood glucose in plasma was measured by using an automated analyzer (YSI 2300 STAT Plus). Blood cell counts were measured by a hematology automated analyzer (Sysmex).

Biochemical analysis

The following assays were completed in-house at Victoria University. Plasma insulin concentration was determined by RIA according to the manufacturer’s instructions with an intravariability and intervariability of 4.2% and 3.8%, respectively (HI-14K, EMD Merck Millipore). Results from the OGTT were used to determine both insulin sensitivity and resistance using area under the curve for plasma insulin and glucose concentrations and the homeostatic assessment indices of insulin sensitivity calculated as (fasting insulin × fasting glucose/22.5) (35). IL-6 was measured using a high-sensitivity ELISA with an intraassay variability of 7.6% (ab46042, Abcam). S-Adenosylmethionine (SAM) was determined by competitive enzyme immunoassay and had an intraassay variability of 8.8% (STA-672, Cell Biolabs).

The following assays were completed in the accredited pathology laboratory at Monash Health, Australia. Lipid profiles (cholesterol, triglycerides, low-density lipoprotein, and high-density lipoprotein) were quantified by automated enzymatic methods (Architect C18000 analyzer), and high-sensitivity C-reactive protein was quantified by a highly sensitive near-infrared particle immunoassay using the Synchron LX system chemistry analyzer (Beckman Coulter). Folate was determined by the Access/Dxi folate assay, which is a competitive binding receptor assay performed on the Unicel DXI 800 system (Beckman Coulter). Homocysteine was measured using a Synchron DXC800 system chemistry analyzer (Beckman Coulter). Anti-Müllerian hormone (AMH) was analyzed using an ELISA (A16507, Immunotech/Beckman Coulter). The SHBG assay was performed using a sequential two-step immunoenzymatic (“sandwich”) assay carried out on a Unicel DXI 800 (Beckman Coulter). Testosterone was measured using the HPLC–tandem mass spectrometry method using a liquid sample extraction (AB Sciex Triple Quad 5500 liquid chromatography–tandem mass spectrometry). Free androgen index was calculated as (total testosterone × 100)/SHBG. Free testosterone (fT) was calculated by the Södergard fT calculation (36).

PBMC isolation

PBMCs were isolated by Ficoll gradient as previously described in Simar et al. (28) with some modifications. After centrifugation (1200 × g for 20 minutes, brake off) and removal of plasma, the cell suspension was diluted in RPMI 1640 (Thermo Fisher), supplemented with penicillin/streptomycin and l-glutamine, and carefully layered on Ficoll in Sepmate™ tubes (Stem Cell eTchnologies) in equal volumes and centrifuged. The buffy coat containing PBMCs was collected, washed with RPMI 1640, and resuspended in autologous plasma. Cells were counted on an automated cell counter (TC20™ automated cell counter, Bio-Rad Laboratories) before being frozen in 10% cyropreservant dimethyl sulfoxide in RPMI 1640 overnight in a controlled rate freezing container (CoolCell®, BioCision) and transferred to liquid nitrogen the following day for long-term storage.

Global DNA methylation

Global methylation in PBMCs was quantified as previously described (28). Briefly, the cells were stained with an antibody cocktail of phycoerythrin-conjugated anti-CD3, peridinin chlorophyll–conjugated anti-CD8 and anti-CD14, and allophycocyanin-conjugated anti-CD4 and anti-CD19 (Becton Dickinson). Cells were then further stained with anti–5-methylcytosine (AbD Serotec, Bio-Rad Laboratories) or with its associated isotope control (mouse IgG1, BD Biosciences) labeled with Alexa Fluor 488 according to the manufacturer’s instructions (Zenon Alexa Fluor 488 mouse IgG1, Molecular Probes, Life Technologies). Cells were incubated (20 minutes in the dark at room temperature) and then run immediately on the flow cytometer (FACSCalibur, BD Biosciences). A specific gating strategy (37) was used to separate the different cell populations and the median fluorescence intensity was measured and normalized by the median fluorescence intensity from the isotope controls for each cell population. Data were analyzed using FlowJo version 10 (Tree Star) and Cytobank.

Cell sorting

Cryopreserved PBMCs were thawed and immediately resuspended in RPMI 1640 before being washed and stained with anti-CD3 (allophycocyanin), anti-CD4 (phycoerythrin), anti-CD8 (Brilliant Violet 510), anti-CD19 (Brilliant Violet 421), anti-CD20 (phycoerythrin-cyanine or Cy7), and anti-CD14 (allophycocyanin). All antibodies were sourced from BD Biosciences (37). The stained PBMCs were washed twice in sort buffer [PBS 1% BSA, 25 mM HEPES (pH 7.0), 1 mM EDTA] before being sorted into four populations (monocytes and T helper, T cytotoxic, and B cells) using the FACSAria (BD Biosciences).

RNA sequencing

Total RNA from T helper cells was extracted using the Qiagen AllPrep DNA/RNA/miRNA universal kit (no. 80224, Qiagen) following the manufacturer’s instructions. Quality of RNA was established using the Agilent RNA 600 Nano kit and Bioanalyzer instrument (Agilent Technologies). RNA sequencing was performed according to Illumina TruSeq stranded total RNA with the Ribo-Zero Gold protocol (Illumina) as previously described (38). Each library was quantified to ensure optimum cluster densities across every lane of the flow cell using the Qubit double-stranded DNA HS assay kit (Invitrogen). Quality control for base pair size and purity was assessed using an Agilent high-sensitivity DNA chip and Bioanalyzer instrument (Agilent Technologies). Each library was diluted to 1 nM before being pooled and measured on the Illumina NextSeq 500 (Illumina).

Reduced representation bisulfite sequencing

Genomic DNA was extracted from T helper cells using the Qiagen AllPrep DNA/RNA/miRNA universal kit (no. 80224, Qiagen) following the manufacturer’s instructions. Reduced representation bisulfite sequencing (RRBS) was performed using a premium RRBS kit (no. C02030033, Diagenode) following the manufacturer’s instructions. Each library was quantified using the Qubit double-stranded DNA HS assay kit (Invitrogen) and quality controlled for base pair size and purity using the an Agilent high-sensitivity DNA chip and Bioanalyzer instrument (Agilent Technologies). Genome-wide DNA methylation of T helper cells was measured on the Illumina NextSeq 500 (Illumina).

Transcriptomic analysis

RNA sequencing raw reads were aligned to human genome (hg38) using STAR (39), and gene coverages were computed by featureCounts (40) using Gencode annotation (41). The ribosomal RNA counts were excluded from the downstream analysis. Libraries with <15 million assigned reads were removed from analysis. A generalized linear model (y ∼ 0 + disease) was fitted for disease factor by using DESeq2 pipeline (42). Genes with a false discovery rate <0.1 were considered differentially expressed.

Approximately 15 million reads per sample were assigned to genes, with 38 genes surviving the expression threshold (q < 0.1). Molecular functions and biological processes were established from the Universal Protein Resource (UniProt), which is a comprehensive resource for protein sequence and annotation data. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database investigated the interaction of molecular pathways that were associated with the identified differentially expressed genes. This provided an in silico indication of the molecular pathways that may be affected in the T helper cells of women with PCOS.

RRBS analysis

RRBS reads were processed with the “rrbs” setting of Trim Galore v0.3.7 and Cutadapt v1.4.2. Processed reads were mapped to hg38 followed by derivation of CpG methylation using Bismark (43). Mappings to Y chromosome were removed for the differential methylation analysis. Differential methylation analysis was conducted on site and region level according to the sample groups (PCOS vs control) by using RnBeads pipeline (44). For each library, single-nucleotide polymorphism–enriched sites were removed and sites with <10 counts were masked. Gene ontology (GO) enrichment analysis was conducted to determine significant GO terms using a hypergeometric test and a rank cutoff applied for the top 100 best ranking regions in RnBeads pipeline.

Statistical analysis

Data were analyzed using IBM SPSS Statistics version 22. Baseline characteristics are presented as mean ± SD or median (interquartile range), when data were skewed. The baseline characteristics between PCOS and control groups were compared using Student t tests when data were normally distributed. Data were tested for normality using the Shapiro–Wilk test and when deemed nonnormal the Mann–Whitney test was used to compare the baseline characteristics. Student t tests were used to examine the difference in global DNA methylation between women with and without PCOS. Statistical significance was accepted at P < 0.05. To get insight into the relationship between clinical perturbations and DNA methylation in blood, we performed Spearman correlation analyses in women with or without PCOS for each population of PBMCs, assessing the association between body composition, physical activity, dietary intake, and hormones with global DNA methylation. After adjusting for multiple comparisons, statistical significance was accepted when false discovery rate q was ≤0.1. The data sets generated and/or analyzed during the current study are available in the National Center for Biotechnology Information’s Gene Expression Omnibus database through the Gene Expression Omnibus series accession number GSE130582 (45).

Results

Clinical and biochemical characteristics

All 17 women with PCOS had irregular cycles or amenorrhea; 14 had clinical or biochemical hyperandrogenism, and 15 had polycystic ovaries on ultrasound. Women without PCOS did not display any of the three clinical features detailed by the Rotterdam criteria (32, 46). Detailed clinical characteristics of the 34 women (n = 17 PCOS; n = 17 without PCOS) who participated in study are reported in Table 1. There were no differences in age, height, weight, BMI, markers of body composition, physical activity, energy, and macronutrient intake between women with or without PCOS (Table 1). There were also no differences in the lipid profile, circulating methyl donors (SAM or homocysteine), cytokines (IL or IL-6 and high-sensitivity C-reactive protein), or a difference in the fasting and the postprandial response to the OGTT and homeostatic model assessment for assessing insulin resistance (Table 2). There was a tendency for folate to be higher in women with PCOS (P = 0.08). Women with PCOS showed a higher area under the glucose curve (P = 0.02) during the OGTT compared with women without PCOS, but there was no difference observed for the area under the insulin curve between groups (Table 2). Women with PCOS had higher levels of fT (P < 0.01), free androgen index (P < 0.01), and AMH (P < 0.01) (Table 2).

Table 1.

Anthropometric Data and Lifestyle Characteristics

Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P Value
General characteristics
 Age, y3430.1 ± 6.628.9 ± 4.80.6
 Height, cm34165 ± 7.2162 ± 4.70.3
 Weight, kg3470.5 ± 16.871.5 ± 18.20.9
 BMI, kg/m23425.5 ± 5.426.6 ± 6.90.6
 WHR320.85 (0.8, 1.0)0.80 (0.8, 0.9)0.2
 Body fat, %3132.3 (25.9, 40.5)37.7 (31.8, 41.8)0.3
Physical activity
 PA (IPAQ), min⋅wk31172 (15.0, 427.5)165 (40.0, 255.0)0.6
 PA (accel), min⋅d3047.0 (45.5, 48.5)47.0 (46.0, 50.5)0.6
Food intake
 Energy, g318269 (7033, 11,402)7744 (5509, 8885)0.2
 Saturated fats, g3128.0 (23.0, 50.0)21.5 (17.2, 30.3)0.07
 Carbohydrates, g31198 (157, 278)171 (144, 220)0.2
 Sugar, g3170.0 (56.5, 118)67.5 (51.0, 123)0.6
Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P Value
General characteristics
 Age, y3430.1 ± 6.628.9 ± 4.80.6
 Height, cm34165 ± 7.2162 ± 4.70.3
 Weight, kg3470.5 ± 16.871.5 ± 18.20.9
 BMI, kg/m23425.5 ± 5.426.6 ± 6.90.6
 WHR320.85 (0.8, 1.0)0.80 (0.8, 0.9)0.2
 Body fat, %3132.3 (25.9, 40.5)37.7 (31.8, 41.8)0.3
Physical activity
 PA (IPAQ), min⋅wk31172 (15.0, 427.5)165 (40.0, 255.0)0.6
 PA (accel), min⋅d3047.0 (45.5, 48.5)47.0 (46.0, 50.5)0.6
Food intake
 Energy, g318269 (7033, 11,402)7744 (5509, 8885)0.2
 Saturated fats, g3128.0 (23.0, 50.0)21.5 (17.2, 30.3)0.07
 Carbohydrates, g31198 (157, 278)171 (144, 220)0.2
 Sugar, g3170.0 (56.5, 118)67.5 (51.0, 123)0.6

Abbreviations: Accel, Accelerometer; IPAQ, International Physical Activity Questionnaire; IQR, interquartile range; PA, physical activity; WHR, waist-to-hip ratio.

Table 1.

Anthropometric Data and Lifestyle Characteristics

Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P Value
General characteristics
 Age, y3430.1 ± 6.628.9 ± 4.80.6
 Height, cm34165 ± 7.2162 ± 4.70.3
 Weight, kg3470.5 ± 16.871.5 ± 18.20.9
 BMI, kg/m23425.5 ± 5.426.6 ± 6.90.6
 WHR320.85 (0.8, 1.0)0.80 (0.8, 0.9)0.2
 Body fat, %3132.3 (25.9, 40.5)37.7 (31.8, 41.8)0.3
Physical activity
 PA (IPAQ), min⋅wk31172 (15.0, 427.5)165 (40.0, 255.0)0.6
 PA (accel), min⋅d3047.0 (45.5, 48.5)47.0 (46.0, 50.5)0.6
Food intake
 Energy, g318269 (7033, 11,402)7744 (5509, 8885)0.2
 Saturated fats, g3128.0 (23.0, 50.0)21.5 (17.2, 30.3)0.07
 Carbohydrates, g31198 (157, 278)171 (144, 220)0.2
 Sugar, g3170.0 (56.5, 118)67.5 (51.0, 123)0.6
Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P Value
General characteristics
 Age, y3430.1 ± 6.628.9 ± 4.80.6
 Height, cm34165 ± 7.2162 ± 4.70.3
 Weight, kg3470.5 ± 16.871.5 ± 18.20.9
 BMI, kg/m23425.5 ± 5.426.6 ± 6.90.6
 WHR320.85 (0.8, 1.0)0.80 (0.8, 0.9)0.2
 Body fat, %3132.3 (25.9, 40.5)37.7 (31.8, 41.8)0.3
Physical activity
 PA (IPAQ), min⋅wk31172 (15.0, 427.5)165 (40.0, 255.0)0.6
 PA (accel), min⋅d3047.0 (45.5, 48.5)47.0 (46.0, 50.5)0.6
Food intake
 Energy, g318269 (7033, 11,402)7744 (5509, 8885)0.2
 Saturated fats, g3128.0 (23.0, 50.0)21.5 (17.2, 30.3)0.07
 Carbohydrates, g31198 (157, 278)171 (144, 220)0.2
 Sugar, g3170.0 (56.5, 118)67.5 (51.0, 123)0.6

Abbreviations: Accel, Accelerometer; IPAQ, International Physical Activity Questionnaire; IQR, interquartile range; PA, physical activity; WHR, waist-to-hip ratio.

Table 2.

Metabolic and Reproductive Profile of Participating Women With and Without PCOS

Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P
Lipid profile
 Cholesterol, mmol/L304.2 ± 0.74.4 ± 0.70.5
 Triglycerides, mmol/L300.6 (0.6, 0.9)0.8 (0.5, 1.0)0.6
 HDL, mmol/L301.5 ± 0.31.5 ± 0.31.0
 LDL, mmol/L302.5 ± 0.62.4 ± 0.70.6
OGTT
 Fasting glucose, mmol/L344.9 (4.8, 5.4)4.9 (4.6, 5.3)0.4
 Fasting insulin, pmol/L3472.8 (56.2, 89.4)68.5 (47.2, 102.4)0.9
 2-h Glucose, mmol/L344.75 (4.5, 5.4)5.5 (4.6, 5.8)0.2
 2-h Insulin, pmol/L34278 (207, 399)309 (165, 737)0.5
 AUC glucose, mmol/L33665.8 ± 75.4783.7 ± 167.80.02
 AUC insulin, mmol/L3348,012 ± 18,36766,180 ± 40,3500.11
 HOMA342.65 (2.0, 3.2)2.41 (1.7, 4.4)0.9
Reproductive markers
 fT, pM3423.2 (17.7, 31.2)10.80 (10.3, 14.1)<0.001
 SHBG, nmol/L3462.1 ± 21.563.4 ± 38.40.9
 FAI341.42 (1.4, 2.2)3.17 (2.2, 5.3)<0.001
 AMH, pmol/L3318.9 (10.8, 31.1)48.6 (37.7, 74.6)<0.001
Systemic methyl substrate/donors
 SAM, µg/mL327.7 (5.4, 8.3)6.9 (6.0, 12.8)0.7
 Folate, nmol/L3329.9 (23.2, 37.5)37.3 (23.4, 45.8)0.08
 Homocysteine339.0 (6.9, 10.1)7.1 (6.4, 9.7)0.3
Cytokines
 HsCRP, mg/L301.3 (0.5, 3.3)1.4 (0.6, 4.5)0.7
 IL-6, pg/ml311.8 (1.3, 3.0)2.1 (1.5, 2.9)0.3
Cell counts
 WBC, µL315335 ± 12295750 ± 14970.4
Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P
Lipid profile
 Cholesterol, mmol/L304.2 ± 0.74.4 ± 0.70.5
 Triglycerides, mmol/L300.6 (0.6, 0.9)0.8 (0.5, 1.0)0.6
 HDL, mmol/L301.5 ± 0.31.5 ± 0.31.0
 LDL, mmol/L302.5 ± 0.62.4 ± 0.70.6
OGTT
 Fasting glucose, mmol/L344.9 (4.8, 5.4)4.9 (4.6, 5.3)0.4
 Fasting insulin, pmol/L3472.8 (56.2, 89.4)68.5 (47.2, 102.4)0.9
 2-h Glucose, mmol/L344.75 (4.5, 5.4)5.5 (4.6, 5.8)0.2
 2-h Insulin, pmol/L34278 (207, 399)309 (165, 737)0.5
 AUC glucose, mmol/L33665.8 ± 75.4783.7 ± 167.80.02
 AUC insulin, mmol/L3348,012 ± 18,36766,180 ± 40,3500.11
 HOMA342.65 (2.0, 3.2)2.41 (1.7, 4.4)0.9
Reproductive markers
 fT, pM3423.2 (17.7, 31.2)10.80 (10.3, 14.1)<0.001
 SHBG, nmol/L3462.1 ± 21.563.4 ± 38.40.9
 FAI341.42 (1.4, 2.2)3.17 (2.2, 5.3)<0.001
 AMH, pmol/L3318.9 (10.8, 31.1)48.6 (37.7, 74.6)<0.001
Systemic methyl substrate/donors
 SAM, µg/mL327.7 (5.4, 8.3)6.9 (6.0, 12.8)0.7
 Folate, nmol/L3329.9 (23.2, 37.5)37.3 (23.4, 45.8)0.08
 Homocysteine339.0 (6.9, 10.1)7.1 (6.4, 9.7)0.3
Cytokines
 HsCRP, mg/L301.3 (0.5, 3.3)1.4 (0.6, 4.5)0.7
 IL-6, pg/ml311.8 (1.3, 3.0)2.1 (1.5, 2.9)0.3
Cell counts
 WBC, µL315335 ± 12295750 ± 14970.4

Abbreviations: AUC, area under the curve; HDL, high-density lipoprotein; HOMA, homeostatic model assessment; HsCRP, high-sensitivity C-reactive protein; FAI, Free Androgen Index; IQR, interquartile range; LDL, low-density lipoprotein; WBC, white blood cell.

Table 2.

Metabolic and Reproductive Profile of Participating Women With and Without PCOS

Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P
Lipid profile
 Cholesterol, mmol/L304.2 ± 0.74.4 ± 0.70.5
 Triglycerides, mmol/L300.6 (0.6, 0.9)0.8 (0.5, 1.0)0.6
 HDL, mmol/L301.5 ± 0.31.5 ± 0.31.0
 LDL, mmol/L302.5 ± 0.62.4 ± 0.70.6
OGTT
 Fasting glucose, mmol/L344.9 (4.8, 5.4)4.9 (4.6, 5.3)0.4
 Fasting insulin, pmol/L3472.8 (56.2, 89.4)68.5 (47.2, 102.4)0.9
 2-h Glucose, mmol/L344.75 (4.5, 5.4)5.5 (4.6, 5.8)0.2
 2-h Insulin, pmol/L34278 (207, 399)309 (165, 737)0.5
 AUC glucose, mmol/L33665.8 ± 75.4783.7 ± 167.80.02
 AUC insulin, mmol/L3348,012 ± 18,36766,180 ± 40,3500.11
 HOMA342.65 (2.0, 3.2)2.41 (1.7, 4.4)0.9
Reproductive markers
 fT, pM3423.2 (17.7, 31.2)10.80 (10.3, 14.1)<0.001
 SHBG, nmol/L3462.1 ± 21.563.4 ± 38.40.9
 FAI341.42 (1.4, 2.2)3.17 (2.2, 5.3)<0.001
 AMH, pmol/L3318.9 (10.8, 31.1)48.6 (37.7, 74.6)<0.001
Systemic methyl substrate/donors
 SAM, µg/mL327.7 (5.4, 8.3)6.9 (6.0, 12.8)0.7
 Folate, nmol/L3329.9 (23.2, 37.5)37.3 (23.4, 45.8)0.08
 Homocysteine339.0 (6.9, 10.1)7.1 (6.4, 9.7)0.3
Cytokines
 HsCRP, mg/L301.3 (0.5, 3.3)1.4 (0.6, 4.5)0.7
 IL-6, pg/ml311.8 (1.3, 3.0)2.1 (1.5, 2.9)0.3
Cell counts
 WBC, µL315335 ± 12295750 ± 14970.4
Clinical FeaturesSample Size (n)Without PCOS [Mean ± SD or Median (IQR)]With PCOS [Mean ± SD or Median (IQR)]P
Lipid profile
 Cholesterol, mmol/L304.2 ± 0.74.4 ± 0.70.5
 Triglycerides, mmol/L300.6 (0.6, 0.9)0.8 (0.5, 1.0)0.6
 HDL, mmol/L301.5 ± 0.31.5 ± 0.31.0
 LDL, mmol/L302.5 ± 0.62.4 ± 0.70.6
OGTT
 Fasting glucose, mmol/L344.9 (4.8, 5.4)4.9 (4.6, 5.3)0.4
 Fasting insulin, pmol/L3472.8 (56.2, 89.4)68.5 (47.2, 102.4)0.9
 2-h Glucose, mmol/L344.75 (4.5, 5.4)5.5 (4.6, 5.8)0.2
 2-h Insulin, pmol/L34278 (207, 399)309 (165, 737)0.5
 AUC glucose, mmol/L33665.8 ± 75.4783.7 ± 167.80.02
 AUC insulin, mmol/L3348,012 ± 18,36766,180 ± 40,3500.11
 HOMA342.65 (2.0, 3.2)2.41 (1.7, 4.4)0.9
Reproductive markers
 fT, pM3423.2 (17.7, 31.2)10.80 (10.3, 14.1)<0.001
 SHBG, nmol/L3462.1 ± 21.563.4 ± 38.40.9
 FAI341.42 (1.4, 2.2)3.17 (2.2, 5.3)<0.001
 AMH, pmol/L3318.9 (10.8, 31.1)48.6 (37.7, 74.6)<0.001
Systemic methyl substrate/donors
 SAM, µg/mL327.7 (5.4, 8.3)6.9 (6.0, 12.8)0.7
 Folate, nmol/L3329.9 (23.2, 37.5)37.3 (23.4, 45.8)0.08
 Homocysteine339.0 (6.9, 10.1)7.1 (6.4, 9.7)0.3
Cytokines
 HsCRP, mg/L301.3 (0.5, 3.3)1.4 (0.6, 4.5)0.7
 IL-6, pg/ml311.8 (1.3, 3.0)2.1 (1.5, 2.9)0.3
Cell counts
 WBC, µL315335 ± 12295750 ± 14970.4

Abbreviations: AUC, area under the curve; HDL, high-density lipoprotein; HOMA, homeostatic model assessment; HsCRP, high-sensitivity C-reactive protein; FAI, Free Androgen Index; IQR, interquartile range; LDL, low-density lipoprotein; WBC, white blood cell.

Global DNA methylation

Global DNA methylation analysis of the individual immune cell populations was performed using an antibody that detects methylated cytosine bases coupled with FACS detection (28). We identified hypomethylation in monocytes (P = 0.006) and T helper (P = 0.004), T cytotoxic (P = 0.004), and B cells (P = 0.03) in women with PCOS compared with women without PCOS (Fig. 1). Furthermore, systemic AMH concentration was associated with global DNA hypo-methylation in T helper cells (rs = −0.428, q = 0.078), but not in T cytotoxic cells, B cells, or monocytes (Table 3). Subset analysis revealed that the AMH-associated hypomethylation in T helper cells was only observed in women with PCOS (P = 0.035). fT was also associated with DNA hypomethylation in T helper cells (rs = −0.381, q < 0.087), but not T cytotoxic cells, B cells, or monocytes (Table 3). Collectively, our results show a cell type–specific alteration of global DNA methylation in immune cells in PCOS and suggest a correlation with the dysregulated ovarian hormone and hyperandrogenism as indicated by associations with AMH and fT, respectively.

Lower 5-methylcytosine levels in monocytes and T helper, T cytotoxic, and B cells in women with PCOS. Comparison of 5-methylcytosine median fluorescence intensity (MFI) between women with PCOS (square) and women without PCOS (circle). MFI was normalized by the MFI from the isotope control (CON). *P < 0.05, **P < 0.01, vs control.
Figure 1.

Lower 5-methylcytosine levels in monocytes and T helper, T cytotoxic, and B cells in women with PCOS. Comparison of 5-methylcytosine median fluorescence intensity (MFI) between women with PCOS (square) and women without PCOS (circle). MFI was normalized by the MFI from the isotope control (CON). *P < 0.05, **P < 0.01, vs control.

Table 3.

Spearman Correlations of a priori Covariates With Global DNA Methylation in T Helper Cells, T Cytotoxic Cells, Monocytes, and B Cells Across All Participants

CovariateSpearman Correlation
T Helper CellsT Cytotoxic CellsMonocytesB Cells
BMI, kg/m2rs0.0070.0210.1400.143
P value0.9670.9050.4300.420
FDR q value0.9670.9000.5160.504
PA (MVPA), min⋅drs−0.139−0.166−0.165−0.213
P value0.4630.3800.3830.259
FDR q value0.6940.7350.51640.504
Energy, grs0.070−0.068−0.069−0.095
P value0.7080.7160.7120.612
FDR q value0.8460.8520.85460.612
AMH, pmol/Lrs−0.428−0.317−0.286−0.191
P value0.0130.0720.1060.288
FDR q value0.0780.4320.3510.504
fT, pMrs−0.381−0.237−0.278−0.175
P value0.0290.1830.1170.330
FDR q value0.0870.5490.3510.504
AUC glucose, mmol/Lrs−0.160−0.124−0.152−0.154
P value0.3730.4930.3990.393
FDR q value0.6940.7350.5160.504
CovariateSpearman Correlation
T Helper CellsT Cytotoxic CellsMonocytesB Cells
BMI, kg/m2rs0.0070.0210.1400.143
P value0.9670.9050.4300.420
FDR q value0.9670.9000.5160.504
PA (MVPA), min⋅drs−0.139−0.166−0.165−0.213
P value0.4630.3800.3830.259
FDR q value0.6940.7350.51640.504
Energy, grs0.070−0.068−0.069−0.095
P value0.7080.7160.7120.612
FDR q value0.8460.8520.85460.612
AMH, pmol/Lrs−0.428−0.317−0.286−0.191
P value0.0130.0720.1060.288
FDR q value0.0780.4320.3510.504
fT, pMrs−0.381−0.237−0.278−0.175
P value0.0290.1830.1170.330
FDR q value0.0870.5490.3510.504
AUC glucose, mmol/Lrs−0.160−0.124−0.152−0.154
P value0.3730.4930.3990.393
FDR q value0.6940.7350.5160.504

Significance was considered with a false discovery rate q of <0.1. Bold text indicates statistical significance.

Abbreviations: AUC, area under the curve; FDR, false discovery rate; MVPA, moderate vigorous physical activity; PA; physical activity; rs, Spearman Rho.

Table 3.

Spearman Correlations of a priori Covariates With Global DNA Methylation in T Helper Cells, T Cytotoxic Cells, Monocytes, and B Cells Across All Participants

CovariateSpearman Correlation
T Helper CellsT Cytotoxic CellsMonocytesB Cells
BMI, kg/m2rs0.0070.0210.1400.143
P value0.9670.9050.4300.420
FDR q value0.9670.9000.5160.504
PA (MVPA), min⋅drs−0.139−0.166−0.165−0.213
P value0.4630.3800.3830.259
FDR q value0.6940.7350.51640.504
Energy, grs0.070−0.068−0.069−0.095
P value0.7080.7160.7120.612
FDR q value0.8460.8520.85460.612
AMH, pmol/Lrs−0.428−0.317−0.286−0.191
P value0.0130.0720.1060.288
FDR q value0.0780.4320.3510.504
fT, pMrs−0.381−0.237−0.278−0.175
P value0.0290.1830.1170.330
FDR q value0.0870.5490.3510.504
AUC glucose, mmol/Lrs−0.160−0.124−0.152−0.154
P value0.3730.4930.3990.393
FDR q value0.6940.7350.5160.504
CovariateSpearman Correlation
T Helper CellsT Cytotoxic CellsMonocytesB Cells
BMI, kg/m2rs0.0070.0210.1400.143
P value0.9670.9050.4300.420
FDR q value0.9670.9000.5160.504
PA (MVPA), min⋅drs−0.139−0.166−0.165−0.213
P value0.4630.3800.3830.259
FDR q value0.6940.7350.51640.504
Energy, grs0.070−0.068−0.069−0.095
P value0.7080.7160.7120.612
FDR q value0.8460.8520.85460.612
AMH, pmol/Lrs−0.428−0.317−0.286−0.191
P value0.0130.0720.1060.288
FDR q value0.0780.4320.3510.504
fT, pMrs−0.381−0.237−0.278−0.175
P value0.0290.1830.1170.330
FDR q value0.0870.5490.3510.504
AUC glucose, mmol/Lrs−0.160−0.124−0.152−0.154
P value0.3730.4930.3990.393
FDR q value0.6940.7350.5160.504

Significance was considered with a false discovery rate q of <0.1. Bold text indicates statistical significance.

Abbreviations: AUC, area under the curve; FDR, false discovery rate; MVPA, moderate vigorous physical activity; PA; physical activity; rs, Spearman Rho.

DNA methylome of T helper cells

To further explore the changes in the methylome of immune cells from women with PCOS, we investigated genome-wide methylation levels in sorted T helper cells. We identified 5581 CpG sites differentially methylated in women with PCOS in T helper cells. Of these sites, only 13 were located within promoter regions or gene bodies (8 promoters and 5 gene bodies, Table 4). Gene enrichment analysis of the genes harboring differentially methylated cytosines identified GO terms related to T cell function, as well as reproductive function, including female pregnancy, response to prolactin, regulation of ovarian follicle development, progesterone receptor signaling pathway, male sex determination, and response to steroid hormone (37). Thus, our results indicate that genes regulating reproductive function are epigenetically remodeled at in specific immune cells.

Table 4.

Genes and Promoters Carrying Differentially Methylated CpGs in T Helper Cells From Women With PCOS Compared With Women Without PCOS

Gene SymbolGene NameENSEMBL Gene IDChromosomeFunction and Gene Type
Genes
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 AC025678.3N/AENSG00000279092chr15To be experimentally confirmed protein tyrosine kinase involved in T cell signaling and activation
 APP006565.1N/AENSG00000279092chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
Promoter of genes
 WBP11P1WW domain binding protein 11 pseudogene 1ENSG00000260389chr2Pseudogene
 SCGB3A2Secretoglobin family 3A member 2ENSG00000164265chr5Receptor-mediated endocytosis
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 OVCH1Ovochymase 1ENSG00000187950chr12Serine-type endopeptidase activity, hydrolase activity, metal ion binding, proteolysis
 AP006565.1N/AENSG00000265737chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
 IGLJ2Immunoglobulin lambda joining 2ENSG00000211676chr22Immunoglobin
Gene SymbolGene NameENSEMBL Gene IDChromosomeFunction and Gene Type
Genes
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 AC025678.3N/AENSG00000279092chr15To be experimentally confirmed protein tyrosine kinase involved in T cell signaling and activation
 APP006565.1N/AENSG00000279092chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
Promoter of genes
 WBP11P1WW domain binding protein 11 pseudogene 1ENSG00000260389chr2Pseudogene
 SCGB3A2Secretoglobin family 3A member 2ENSG00000164265chr5Receptor-mediated endocytosis
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 OVCH1Ovochymase 1ENSG00000187950chr12Serine-type endopeptidase activity, hydrolase activity, metal ion binding, proteolysis
 AP006565.1N/AENSG00000265737chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
 IGLJ2Immunoglobulin lambda joining 2ENSG00000211676chr22Immunoglobin

Abbreviation: N/A, not applicable.

Table 4.

Genes and Promoters Carrying Differentially Methylated CpGs in T Helper Cells From Women With PCOS Compared With Women Without PCOS

Gene SymbolGene NameENSEMBL Gene IDChromosomeFunction and Gene Type
Genes
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 AC025678.3N/AENSG00000279092chr15To be experimentally confirmed protein tyrosine kinase involved in T cell signaling and activation
 APP006565.1N/AENSG00000279092chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
Promoter of genes
 WBP11P1WW domain binding protein 11 pseudogene 1ENSG00000260389chr2Pseudogene
 SCGB3A2Secretoglobin family 3A member 2ENSG00000164265chr5Receptor-mediated endocytosis
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 OVCH1Ovochymase 1ENSG00000187950chr12Serine-type endopeptidase activity, hydrolase activity, metal ion binding, proteolysis
 AP006565.1N/AENSG00000265737chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
 IGLJ2Immunoglobulin lambda joining 2ENSG00000211676chr22Immunoglobin
Gene SymbolGene NameENSEMBL Gene IDChromosomeFunction and Gene Type
Genes
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 AC025678.3N/AENSG00000279092chr15To be experimentally confirmed protein tyrosine kinase involved in T cell signaling and activation
 APP006565.1N/AENSG00000279092chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
Promoter of genes
 WBP11P1WW domain binding protein 11 pseudogene 1ENSG00000260389chr2Pseudogene
 SCGB3A2Secretoglobin family 3A member 2ENSG00000164265chr5Receptor-mediated endocytosis
 COX6CP15Cytochrome c oxidase subunit 6C pseudogene 15ENSG00000228092chr10Pseudogene
 SCGB1D4Secretoglobin family 1D member 4ENSG00000197745chr11Regulation of chemotactic cell migration and invasion
 OVCH1Ovochymase 1ENSG00000187950chr12Serine-type endopeptidase activity, hydrolase activity, metal ion binding, proteolysis
 AP006565.1N/AENSG00000265737chr18Antisense RNA
 AC104301.2N/AN/Achr20Noncoding RNA
 IGLJ2Immunoglobulin lambda joining 2ENSG00000211676chr22Immunoglobin

Abbreviation: N/A, not applicable.

Transcriptomic analysis of T helper cells

To determine whether the specific DNA methylation signature we identified in T helper cells is linked to transcriptional changes, we investigated gene expression via RNA sequencing. We found 37 genes differentially expressed between women with PCOS (Fig. 2) compared with those without PCOS, with 33 downregulated and 4 upregulated (Table 5). Surprisingly, none of the differentially expressed genes showed changes in DNA methylation as identified by RRBS analysis. Interestingly a large portion of the differentially expressed transcripts (n = 17) were long noncoding RNAs (lncRNA), including pseudogenes, long intergenic noncoding RNA, and Y RNA. The remaining 20 transcripts were protein coding and related to inflammatory and immune cell function. KEGG analysis revealed that the cyclic guanosine monophosphate (cGMP) signaling pathway and bone morphogenetic pathway (BMP) signaling pathway were functionally enriched. BMP signaling was upregulated, whereas cGMP signaling was downregulated, in women with PCOS. Taken together, our results indicate that epigenetic remodeling of genes related to reproductive function in T helper cells does not seem to be functional at the transcriptomic level in women with PCOS.

Volcano plots representing differentially expressed genes in T helper cells of women with PCOS compared with women without PCOS. Red circles highlight differentially expressed genes. False discovery rate q of <0.1. padj, adjusted P value.
Figure 2.

Volcano plots representing differentially expressed genes in T helper cells of women with PCOS compared with women without PCOS. Red circles highlight differentially expressed genes. False discovery rate q of <0.1. padj, adjusted P value.

Table 5.

Differentially Expressed Genes in T Helper Cells From Women With PCOS Compared With Women Without PCOS

Gene NameGene SymbolENSEMBL Gene IDLog2 Fold ChangePadjGene TypeMolecular FunctionMolecular Process
Downregulated genes in women with PCOS
 N/AAC138969.2ENSG00000277920−7.70.025PseudogeneUnknownUnknown
 Ring finger protein 217RNF217ENSG00000146373−7.70.009Protein codingMetal ion binding, ubiquitin-protein transferase activityUnknown
 FKBP prolyl isomerase 1BFKBP1BENSG00000119782−7.30.029Protein codingPeptidyl-prolyl cis-trans isomerase activityUnknown
 Guanylate cyclase 1 soluble subunit alpha 2GUCY1A2ENSG00000152402−7.10.062Protein codingGTP binding, phosphorus-oxygen lyase activity guanylate cyclase activity, heme bindingIntracellular signal transduction, cyclic nucleotide biosynthetic process, positive regulation of cGMP biosynthetic process, adenylate cyclase activity
 Caveolae associated protein 2CAVIN2ENSG00000168497−6.80.037Protein codingPhosphatidylserine binding, phospholipid binding, protein kinase C bindingPlasma membrane tubulation
 Alcohol dehydrogenase 4 (class II), pi polypeptideADH4ENSG00000198099−6.60.062Protein codingAlcohol dehydrogenase (NAD) activity, all-trans retinal binding, ethanol binding, NAD bindingAlcohol catabolic process, cellular aldehyde metabolic process, ethanol oxidation
 Solute carrier family 5 member 11SLC5A11ENSG00000158865−6.60.025Protein codingTransporter activityTransmembrane transport
 VWA8 antisense RNA 1 (head to head)VWA8-AS1ENSG00000278338−6.50.025lncRNAUnknownUnknown
 N/AZ98752.2ENSG00000234271−6.50.071Processed pseudogeneUnknownUnknown
 N/AAC034102.3ENSG00000257449−6.50.044lncRNAUnknownUnknown
 N/AAC253536.6ENSG00000272787.1−6.40.009lincRNAUnknownUnknown
 Atypical chemokine receptor 2ACKR2ENSG00000144648−6.40.074Protein codingC-C chemokine receptor activity, scavenger receptor activity, G-protein–coupled receptor activityChemotaxis, inflammatory response, receptor-mediated endocytosis
 Keratin 74KRT74ENSG00000170484−6.30.078Protein codingKeratin filament binding, structural molecule activityIntermediate filament cytoskeleton organization, cornification, keratinization
 N/ARF00019ENSG00000252652.1−6.30.009Y RNAUnknownUnknown
 Protocadherin gamma subfamily B, 8 pseudogenePCDHGB8PENSG00000248449−6.30.070Transcribed pseudogeneUnknownUnknown
 N/ALOC101928238ENSG00000278611−6.30.074lincRNAUnknownUnknown
 Transmembrane and coiled-coil domains 2TMCO2ENSG00000188800−6.20.058Protein codingUnknownUnknown
 CELF2 antisense RNA 2CELF2-AS2ENSG00000237986−6.20.052lncRNAUnknownUnknown
 N/AAC013476.1ENSG00000234193.1−6.10.025lncRNAUnknownUnknown
 Pannexin 3PANX3ENSG00000154143−5.90.074Protein codingWide pore channel activity, gap junction hemichannel activityCell–cell signaling, transmembrane transport, cation transport
 N/ARF00019ENSG00000202222.1−5.90.029Y RNAUnknownUnknown
 N/ALOC101927851ENSG00000238005−5.20.088lincRNAUnknownUnknown
 Leucine-rich repeat containing 9LRRC9ENSG00000131951−5.20.062Protein codingUnknownUnknown
 EXTL3 antisense RNA 1EXTL3-AS1ENSG00000246339−5.10.040incRNAUnknownUnknown
 KIAA2012KIAA2012ENSG00000182329−4.60.083Protein codingUnknownUnknown
 N/AAC013643.2ENSG00000253875−4.60.062lncRNAUnknownUnknown
 Cytochrome b reductase 1CYBRD1ENSG00000071967−4.40.052Protein codingFerric-chelate reductase activity, protein binding, oxidoreductase activity, oxidizing metal ionsCellular iron ion homeostasis, response to iron ion, oxidation-reduction process
 N/AAC068620.2ENSG00000270147−4.10.078lincRNAUnknownUnknown
 N/AAC243428.1ENSG00000229979−4.10.037Processed pseudogeneUnknownUnknown
 Stearoyl-CoA desaturaseSCDENSG00000099194−2.70.058Protein codingStearoyl-CoA 9-desaturase activity, iron ion binding, oxidoreductase activityLipid metabolic process, fatty acid biosynthetic process, regulation of cholesterol biosynthetic process, oxidation-reduction process
 NIMA related kinase 10NEK10ENSG00000163491−2.20.061Protein codingNucleotide binding, protein serine/threonine kinase activity, ATP binding, transferase activityProtein phosphorylation, positive regulation of protein autophosphorylation, positive regulation of MAPK activity, regulation of ERK1 and ERK2 cascade, regulation of cell cycle G2/M phase transition
 Thymocyte selection associated family member 2THEMIS2ENSG00000130775−0.90.009Protein codingProtein bindingImmune system process, inflammatory response, cell adhesion, T cell receptor signaling pathway
 cAMP-dependent protein kinase inhibitor alphaPKIAENSG00000171033−0.50.083Protein codingProtein kinase inhibitor activity, cAMP-dependent protein kinase inhibitor activity,Negative regulation of transcription by RNA polymerase II, negative regulation of protein kinase activity, regulation of G2/M transition of mitotic cell cycle, negative regulation of protein import into nucleus, negative regulation of catalytic activity
Upregulated genes in women with PCOS
 MX dynamin like GTPase 2MX2ENSG000001834860.60.0711Protein codingNucleotide binding, GTPase activity, protein binding, microtubule bindingMitochondrial fission, immune system process, defense response, response to virus, response to interferon-α, innate immune response, mRNA transport
 Small Cajal body–specific RNA 21SCARNA21ENSG000002528350.60.082652scaRNAUnknownUnknown
 Histone cluster 1 H3 family member cHIST1H3CENSG000002782721.40.073504Protein codingDNA binding, protein binding, cadherin binding, protein heterodimerization activityChromatin silencing at rDNA, nucleosome assembly, telomere organization, IL-7–mediated signaling pathway, negative regulation of gene expression, epigenetic, regulation of hematopoietic stem cell differentiation, regulation of gene silencing
 SMAD family member 1SMAD1ENSG000001703653.00.099453Protein codingRNA polymerase II proximal promoter sequence-specific DNA binding, signal transducer activity, downstream of receptor, TGF-β receptor, pathway-specific cytoplasmic mediator activity, protein homodimerization activity, co-SMAD binding, I-SMAD binding, primary miRNA bindingMAPK cascade, mesodermal cell fate commitment, osteoblast fate commitment, inflammatory response, TGF-β receptor signaling pathway, SMAD protein complex assembly, gamete generation, embryonic pattern specification, BMP signaling pathway
Gene NameGene SymbolENSEMBL Gene IDLog2 Fold ChangePadjGene TypeMolecular FunctionMolecular Process
Downregulated genes in women with PCOS
 N/AAC138969.2ENSG00000277920−7.70.025PseudogeneUnknownUnknown
 Ring finger protein 217RNF217ENSG00000146373−7.70.009Protein codingMetal ion binding, ubiquitin-protein transferase activityUnknown
 FKBP prolyl isomerase 1BFKBP1BENSG00000119782−7.30.029Protein codingPeptidyl-prolyl cis-trans isomerase activityUnknown
 Guanylate cyclase 1 soluble subunit alpha 2GUCY1A2ENSG00000152402−7.10.062Protein codingGTP binding, phosphorus-oxygen lyase activity guanylate cyclase activity, heme bindingIntracellular signal transduction, cyclic nucleotide biosynthetic process, positive regulation of cGMP biosynthetic process, adenylate cyclase activity
 Caveolae associated protein 2CAVIN2ENSG00000168497−6.80.037Protein codingPhosphatidylserine binding, phospholipid binding, protein kinase C bindingPlasma membrane tubulation
 Alcohol dehydrogenase 4 (class II), pi polypeptideADH4ENSG00000198099−6.60.062Protein codingAlcohol dehydrogenase (NAD) activity, all-trans retinal binding, ethanol binding, NAD bindingAlcohol catabolic process, cellular aldehyde metabolic process, ethanol oxidation
 Solute carrier family 5 member 11SLC5A11ENSG00000158865−6.60.025Protein codingTransporter activityTransmembrane transport
 VWA8 antisense RNA 1 (head to head)VWA8-AS1ENSG00000278338−6.50.025lncRNAUnknownUnknown
 N/AZ98752.2ENSG00000234271−6.50.071Processed pseudogeneUnknownUnknown
 N/AAC034102.3ENSG00000257449−6.50.044lncRNAUnknownUnknown
 N/AAC253536.6ENSG00000272787.1−6.40.009lincRNAUnknownUnknown
 Atypical chemokine receptor 2ACKR2ENSG00000144648−6.40.074Protein codingC-C chemokine receptor activity, scavenger receptor activity, G-protein–coupled receptor activityChemotaxis, inflammatory response, receptor-mediated endocytosis
 Keratin 74KRT74ENSG00000170484−6.30.078Protein codingKeratin filament binding, structural molecule activityIntermediate filament cytoskeleton organization, cornification, keratinization
 N/ARF00019ENSG00000252652.1−6.30.009Y RNAUnknownUnknown
 Protocadherin gamma subfamily B, 8 pseudogenePCDHGB8PENSG00000248449−6.30.070Transcribed pseudogeneUnknownUnknown
 N/ALOC101928238ENSG00000278611−6.30.074lincRNAUnknownUnknown
 Transmembrane and coiled-coil domains 2TMCO2ENSG00000188800−6.20.058Protein codingUnknownUnknown
 CELF2 antisense RNA 2CELF2-AS2ENSG00000237986−6.20.052lncRNAUnknownUnknown
 N/AAC013476.1ENSG00000234193.1−6.10.025lncRNAUnknownUnknown
 Pannexin 3PANX3ENSG00000154143−5.90.074Protein codingWide pore channel activity, gap junction hemichannel activityCell–cell signaling, transmembrane transport, cation transport
 N/ARF00019ENSG00000202222.1−5.90.029Y RNAUnknownUnknown
 N/ALOC101927851ENSG00000238005−5.20.088lincRNAUnknownUnknown
 Leucine-rich repeat containing 9LRRC9ENSG00000131951−5.20.062Protein codingUnknownUnknown
 EXTL3 antisense RNA 1EXTL3-AS1ENSG00000246339−5.10.040incRNAUnknownUnknown
 KIAA2012KIAA2012ENSG00000182329−4.60.083Protein codingUnknownUnknown
 N/AAC013643.2ENSG00000253875−4.60.062lncRNAUnknownUnknown
 Cytochrome b reductase 1CYBRD1ENSG00000071967−4.40.052Protein codingFerric-chelate reductase activity, protein binding, oxidoreductase activity, oxidizing metal ionsCellular iron ion homeostasis, response to iron ion, oxidation-reduction process
 N/AAC068620.2ENSG00000270147−4.10.078lincRNAUnknownUnknown
 N/AAC243428.1ENSG00000229979−4.10.037Processed pseudogeneUnknownUnknown
 Stearoyl-CoA desaturaseSCDENSG00000099194−2.70.058Protein codingStearoyl-CoA 9-desaturase activity, iron ion binding, oxidoreductase activityLipid metabolic process, fatty acid biosynthetic process, regulation of cholesterol biosynthetic process, oxidation-reduction process
 NIMA related kinase 10NEK10ENSG00000163491−2.20.061Protein codingNucleotide binding, protein serine/threonine kinase activity, ATP binding, transferase activityProtein phosphorylation, positive regulation of protein autophosphorylation, positive regulation of MAPK activity, regulation of ERK1 and ERK2 cascade, regulation of cell cycle G2/M phase transition
 Thymocyte selection associated family member 2THEMIS2ENSG00000130775−0.90.009Protein codingProtein bindingImmune system process, inflammatory response, cell adhesion, T cell receptor signaling pathway
 cAMP-dependent protein kinase inhibitor alphaPKIAENSG00000171033−0.50.083Protein codingProtein kinase inhibitor activity, cAMP-dependent protein kinase inhibitor activity,Negative regulation of transcription by RNA polymerase II, negative regulation of protein kinase activity, regulation of G2/M transition of mitotic cell cycle, negative regulation of protein import into nucleus, negative regulation of catalytic activity
Upregulated genes in women with PCOS
 MX dynamin like GTPase 2MX2ENSG000001834860.60.0711Protein codingNucleotide binding, GTPase activity, protein binding, microtubule bindingMitochondrial fission, immune system process, defense response, response to virus, response to interferon-α, innate immune response, mRNA transport
 Small Cajal body–specific RNA 21SCARNA21ENSG000002528350.60.082652scaRNAUnknownUnknown
 Histone cluster 1 H3 family member cHIST1H3CENSG000002782721.40.073504Protein codingDNA binding, protein binding, cadherin binding, protein heterodimerization activityChromatin silencing at rDNA, nucleosome assembly, telomere organization, IL-7–mediated signaling pathway, negative regulation of gene expression, epigenetic, regulation of hematopoietic stem cell differentiation, regulation of gene silencing
 SMAD family member 1SMAD1ENSG000001703653.00.099453Protein codingRNA polymerase II proximal promoter sequence-specific DNA binding, signal transducer activity, downstream of receptor, TGF-β receptor, pathway-specific cytoplasmic mediator activity, protein homodimerization activity, co-SMAD binding, I-SMAD binding, primary miRNA bindingMAPK cascade, mesodermal cell fate commitment, osteoblast fate commitment, inflammatory response, TGF-β receptor signaling pathway, SMAD protein complex assembly, gamete generation, embryonic pattern specification, BMP signaling pathway

Molecular functions and biological processes were established from the Universal Protein Resource (UniProt). False discovery rate q <0.1.

Abbreviations: CoA, conenzyme A; lincRNA, long intergenic noncoding RNA; N/A, not applicable; Padj, adjusted P value; scaRNA, small Cajal body–specific RNA.

Table 5.

Differentially Expressed Genes in T Helper Cells From Women With PCOS Compared With Women Without PCOS

Gene NameGene SymbolENSEMBL Gene IDLog2 Fold ChangePadjGene TypeMolecular FunctionMolecular Process
Downregulated genes in women with PCOS
 N/AAC138969.2ENSG00000277920−7.70.025PseudogeneUnknownUnknown
 Ring finger protein 217RNF217ENSG00000146373−7.70.009Protein codingMetal ion binding, ubiquitin-protein transferase activityUnknown
 FKBP prolyl isomerase 1BFKBP1BENSG00000119782−7.30.029Protein codingPeptidyl-prolyl cis-trans isomerase activityUnknown
 Guanylate cyclase 1 soluble subunit alpha 2GUCY1A2ENSG00000152402−7.10.062Protein codingGTP binding, phosphorus-oxygen lyase activity guanylate cyclase activity, heme bindingIntracellular signal transduction, cyclic nucleotide biosynthetic process, positive regulation of cGMP biosynthetic process, adenylate cyclase activity
 Caveolae associated protein 2CAVIN2ENSG00000168497−6.80.037Protein codingPhosphatidylserine binding, phospholipid binding, protein kinase C bindingPlasma membrane tubulation
 Alcohol dehydrogenase 4 (class II), pi polypeptideADH4ENSG00000198099−6.60.062Protein codingAlcohol dehydrogenase (NAD) activity, all-trans retinal binding, ethanol binding, NAD bindingAlcohol catabolic process, cellular aldehyde metabolic process, ethanol oxidation
 Solute carrier family 5 member 11SLC5A11ENSG00000158865−6.60.025Protein codingTransporter activityTransmembrane transport
 VWA8 antisense RNA 1 (head to head)VWA8-AS1ENSG00000278338−6.50.025lncRNAUnknownUnknown
 N/AZ98752.2ENSG00000234271−6.50.071Processed pseudogeneUnknownUnknown
 N/AAC034102.3ENSG00000257449−6.50.044lncRNAUnknownUnknown
 N/AAC253536.6ENSG00000272787.1−6.40.009lincRNAUnknownUnknown
 Atypical chemokine receptor 2ACKR2ENSG00000144648−6.40.074Protein codingC-C chemokine receptor activity, scavenger receptor activity, G-protein–coupled receptor activityChemotaxis, inflammatory response, receptor-mediated endocytosis
 Keratin 74KRT74ENSG00000170484−6.30.078Protein codingKeratin filament binding, structural molecule activityIntermediate filament cytoskeleton organization, cornification, keratinization
 N/ARF00019ENSG00000252652.1−6.30.009Y RNAUnknownUnknown
 Protocadherin gamma subfamily B, 8 pseudogenePCDHGB8PENSG00000248449−6.30.070Transcribed pseudogeneUnknownUnknown
 N/ALOC101928238ENSG00000278611−6.30.074lincRNAUnknownUnknown
 Transmembrane and coiled-coil domains 2TMCO2ENSG00000188800−6.20.058Protein codingUnknownUnknown
 CELF2 antisense RNA 2CELF2-AS2ENSG00000237986−6.20.052lncRNAUnknownUnknown
 N/AAC013476.1ENSG00000234193.1−6.10.025lncRNAUnknownUnknown
 Pannexin 3PANX3ENSG00000154143−5.90.074Protein codingWide pore channel activity, gap junction hemichannel activityCell–cell signaling, transmembrane transport, cation transport
 N/ARF00019ENSG00000202222.1−5.90.029Y RNAUnknownUnknown
 N/ALOC101927851ENSG00000238005−5.20.088lincRNAUnknownUnknown
 Leucine-rich repeat containing 9LRRC9ENSG00000131951−5.20.062Protein codingUnknownUnknown
 EXTL3 antisense RNA 1EXTL3-AS1ENSG00000246339−5.10.040incRNAUnknownUnknown
 KIAA2012KIAA2012ENSG00000182329−4.60.083Protein codingUnknownUnknown
 N/AAC013643.2ENSG00000253875−4.60.062lncRNAUnknownUnknown
 Cytochrome b reductase 1CYBRD1ENSG00000071967−4.40.052Protein codingFerric-chelate reductase activity, protein binding, oxidoreductase activity, oxidizing metal ionsCellular iron ion homeostasis, response to iron ion, oxidation-reduction process
 N/AAC068620.2ENSG00000270147−4.10.078lincRNAUnknownUnknown
 N/AAC243428.1ENSG00000229979−4.10.037Processed pseudogeneUnknownUnknown
 Stearoyl-CoA desaturaseSCDENSG00000099194−2.70.058Protein codingStearoyl-CoA 9-desaturase activity, iron ion binding, oxidoreductase activityLipid metabolic process, fatty acid biosynthetic process, regulation of cholesterol biosynthetic process, oxidation-reduction process
 NIMA related kinase 10NEK10ENSG00000163491−2.20.061Protein codingNucleotide binding, protein serine/threonine kinase activity, ATP binding, transferase activityProtein phosphorylation, positive regulation of protein autophosphorylation, positive regulation of MAPK activity, regulation of ERK1 and ERK2 cascade, regulation of cell cycle G2/M phase transition
 Thymocyte selection associated family member 2THEMIS2ENSG00000130775−0.90.009Protein codingProtein bindingImmune system process, inflammatory response, cell adhesion, T cell receptor signaling pathway
 cAMP-dependent protein kinase inhibitor alphaPKIAENSG00000171033−0.50.083Protein codingProtein kinase inhibitor activity, cAMP-dependent protein kinase inhibitor activity,Negative regulation of transcription by RNA polymerase II, negative regulation of protein kinase activity, regulation of G2/M transition of mitotic cell cycle, negative regulation of protein import into nucleus, negative regulation of catalytic activity
Upregulated genes in women with PCOS
 MX dynamin like GTPase 2MX2ENSG000001834860.60.0711Protein codingNucleotide binding, GTPase activity, protein binding, microtubule bindingMitochondrial fission, immune system process, defense response, response to virus, response to interferon-α, innate immune response, mRNA transport
 Small Cajal body–specific RNA 21SCARNA21ENSG000002528350.60.082652scaRNAUnknownUnknown
 Histone cluster 1 H3 family member cHIST1H3CENSG000002782721.40.073504Protein codingDNA binding, protein binding, cadherin binding, protein heterodimerization activityChromatin silencing at rDNA, nucleosome assembly, telomere organization, IL-7–mediated signaling pathway, negative regulation of gene expression, epigenetic, regulation of hematopoietic stem cell differentiation, regulation of gene silencing
 SMAD family member 1SMAD1ENSG000001703653.00.099453Protein codingRNA polymerase II proximal promoter sequence-specific DNA binding, signal transducer activity, downstream of receptor, TGF-β receptor, pathway-specific cytoplasmic mediator activity, protein homodimerization activity, co-SMAD binding, I-SMAD binding, primary miRNA bindingMAPK cascade, mesodermal cell fate commitment, osteoblast fate commitment, inflammatory response, TGF-β receptor signaling pathway, SMAD protein complex assembly, gamete generation, embryonic pattern specification, BMP signaling pathway
Gene NameGene SymbolENSEMBL Gene IDLog2 Fold ChangePadjGene TypeMolecular FunctionMolecular Process
Downregulated genes in women with PCOS
 N/AAC138969.2ENSG00000277920−7.70.025PseudogeneUnknownUnknown
 Ring finger protein 217RNF217ENSG00000146373−7.70.009Protein codingMetal ion binding, ubiquitin-protein transferase activityUnknown
 FKBP prolyl isomerase 1BFKBP1BENSG00000119782−7.30.029Protein codingPeptidyl-prolyl cis-trans isomerase activityUnknown
 Guanylate cyclase 1 soluble subunit alpha 2GUCY1A2ENSG00000152402−7.10.062Protein codingGTP binding, phosphorus-oxygen lyase activity guanylate cyclase activity, heme bindingIntracellular signal transduction, cyclic nucleotide biosynthetic process, positive regulation of cGMP biosynthetic process, adenylate cyclase activity
 Caveolae associated protein 2CAVIN2ENSG00000168497−6.80.037Protein codingPhosphatidylserine binding, phospholipid binding, protein kinase C bindingPlasma membrane tubulation
 Alcohol dehydrogenase 4 (class II), pi polypeptideADH4ENSG00000198099−6.60.062Protein codingAlcohol dehydrogenase (NAD) activity, all-trans retinal binding, ethanol binding, NAD bindingAlcohol catabolic process, cellular aldehyde metabolic process, ethanol oxidation
 Solute carrier family 5 member 11SLC5A11ENSG00000158865−6.60.025Protein codingTransporter activityTransmembrane transport
 VWA8 antisense RNA 1 (head to head)VWA8-AS1ENSG00000278338−6.50.025lncRNAUnknownUnknown
 N/AZ98752.2ENSG00000234271−6.50.071Processed pseudogeneUnknownUnknown
 N/AAC034102.3ENSG00000257449−6.50.044lncRNAUnknownUnknown
 N/AAC253536.6ENSG00000272787.1−6.40.009lincRNAUnknownUnknown
 Atypical chemokine receptor 2ACKR2ENSG00000144648−6.40.074Protein codingC-C chemokine receptor activity, scavenger receptor activity, G-protein–coupled receptor activityChemotaxis, inflammatory response, receptor-mediated endocytosis
 Keratin 74KRT74ENSG00000170484−6.30.078Protein codingKeratin filament binding, structural molecule activityIntermediate filament cytoskeleton organization, cornification, keratinization
 N/ARF00019ENSG00000252652.1−6.30.009Y RNAUnknownUnknown
 Protocadherin gamma subfamily B, 8 pseudogenePCDHGB8PENSG00000248449−6.30.070Transcribed pseudogeneUnknownUnknown
 N/ALOC101928238ENSG00000278611−6.30.074lincRNAUnknownUnknown
 Transmembrane and coiled-coil domains 2TMCO2ENSG00000188800−6.20.058Protein codingUnknownUnknown
 CELF2 antisense RNA 2CELF2-AS2ENSG00000237986−6.20.052lncRNAUnknownUnknown
 N/AAC013476.1ENSG00000234193.1−6.10.025lncRNAUnknownUnknown
 Pannexin 3PANX3ENSG00000154143−5.90.074Protein codingWide pore channel activity, gap junction hemichannel activityCell–cell signaling, transmembrane transport, cation transport
 N/ARF00019ENSG00000202222.1−5.90.029Y RNAUnknownUnknown
 N/ALOC101927851ENSG00000238005−5.20.088lincRNAUnknownUnknown
 Leucine-rich repeat containing 9LRRC9ENSG00000131951−5.20.062Protein codingUnknownUnknown
 EXTL3 antisense RNA 1EXTL3-AS1ENSG00000246339−5.10.040incRNAUnknownUnknown
 KIAA2012KIAA2012ENSG00000182329−4.60.083Protein codingUnknownUnknown
 N/AAC013643.2ENSG00000253875−4.60.062lncRNAUnknownUnknown
 Cytochrome b reductase 1CYBRD1ENSG00000071967−4.40.052Protein codingFerric-chelate reductase activity, protein binding, oxidoreductase activity, oxidizing metal ionsCellular iron ion homeostasis, response to iron ion, oxidation-reduction process
 N/AAC068620.2ENSG00000270147−4.10.078lincRNAUnknownUnknown
 N/AAC243428.1ENSG00000229979−4.10.037Processed pseudogeneUnknownUnknown
 Stearoyl-CoA desaturaseSCDENSG00000099194−2.70.058Protein codingStearoyl-CoA 9-desaturase activity, iron ion binding, oxidoreductase activityLipid metabolic process, fatty acid biosynthetic process, regulation of cholesterol biosynthetic process, oxidation-reduction process
 NIMA related kinase 10NEK10ENSG00000163491−2.20.061Protein codingNucleotide binding, protein serine/threonine kinase activity, ATP binding, transferase activityProtein phosphorylation, positive regulation of protein autophosphorylation, positive regulation of MAPK activity, regulation of ERK1 and ERK2 cascade, regulation of cell cycle G2/M phase transition
 Thymocyte selection associated family member 2THEMIS2ENSG00000130775−0.90.009Protein codingProtein bindingImmune system process, inflammatory response, cell adhesion, T cell receptor signaling pathway
 cAMP-dependent protein kinase inhibitor alphaPKIAENSG00000171033−0.50.083Protein codingProtein kinase inhibitor activity, cAMP-dependent protein kinase inhibitor activity,Negative regulation of transcription by RNA polymerase II, negative regulation of protein kinase activity, regulation of G2/M transition of mitotic cell cycle, negative regulation of protein import into nucleus, negative regulation of catalytic activity
Upregulated genes in women with PCOS
 MX dynamin like GTPase 2MX2ENSG000001834860.60.0711Protein codingNucleotide binding, GTPase activity, protein binding, microtubule bindingMitochondrial fission, immune system process, defense response, response to virus, response to interferon-α, innate immune response, mRNA transport
 Small Cajal body–specific RNA 21SCARNA21ENSG000002528350.60.082652scaRNAUnknownUnknown
 Histone cluster 1 H3 family member cHIST1H3CENSG000002782721.40.073504Protein codingDNA binding, protein binding, cadherin binding, protein heterodimerization activityChromatin silencing at rDNA, nucleosome assembly, telomere organization, IL-7–mediated signaling pathway, negative regulation of gene expression, epigenetic, regulation of hematopoietic stem cell differentiation, regulation of gene silencing
 SMAD family member 1SMAD1ENSG000001703653.00.099453Protein codingRNA polymerase II proximal promoter sequence-specific DNA binding, signal transducer activity, downstream of receptor, TGF-β receptor, pathway-specific cytoplasmic mediator activity, protein homodimerization activity, co-SMAD binding, I-SMAD binding, primary miRNA bindingMAPK cascade, mesodermal cell fate commitment, osteoblast fate commitment, inflammatory response, TGF-β receptor signaling pathway, SMAD protein complex assembly, gamete generation, embryonic pattern specification, BMP signaling pathway

Molecular functions and biological processes were established from the Universal Protein Resource (UniProt). False discovery rate q <0.1.

Abbreviations: CoA, conenzyme A; lincRNA, long intergenic noncoding RNA; N/A, not applicable; Padj, adjusted P value; scaRNA, small Cajal body–specific RNA.

Discussion

In this study, we report that immune cells from women with PCOS have a distinct epigenetic profile at both the global and gene-specific levels. On the global level, we found hypomethylation in all PBMC subtypes. In the specific T-lymphocyte population, the differentially methylated genes that we identified were enriched for GO terms related to T cell function, but also reproductive function, suggesting a role of epigenetic reprogramming in the reproductive defects associated with PCOS.

Global DNA methylation

At the cell type–specific level, PBMCs from women with PCOS display distinct global DNA methylation levels compared with women without PCOS. Only one previous study reported that global DNA methylation levels were not different in the total PBMC fraction from women with PCOS (27). Although we have not measured the global methylation of the total PBMC fraction in the current study, differences at the cell type–specific level may not be detected when investigating global DNA methylation at the whole PBMC level. This would be consistent with a previous study showing no difference in global DNA methylation in total blood fractions from subjects with type 2 diabetes whereas global DNA methylation was altered in specific immune cell populations (28), further consolidating the importance of measuring global DNA methylation in immune cells in a cell type–specific manner.

Methylation of DNA is under the control of the one-carbon metabolism, notably the methyl donor SAM, which is a cofactor necessary for the transfer of a methyl group to a cytosine base in DNA (47). To gain insight into the potential relationship between altered global DNA methylation levels in immune cells in PCOS and folate metabolism, we measured key one-carbon metabolites, that is, SAM, homocysteine, and folate, but found no differences. The lack of association between circulating one-carbon metabolites and global DNA methylation levels in immune cells from women with PCOS suggests that methyl donors may not be the primary driver of the global DNA hypomethylation observed in PCOS. Alternatively, it would be interesting to investigate whether the enzymes DNA methyltransferases or ten-eleven translocation, which are responsible for the transfer of the methyl group to the cytosine base or removal of the methyl group, respectively, may play a role in the hypomethylation observed in the immune cell populations (48).

Lifestyle factors such as physical activity and diet have been previously associated with both global and gene-specific DNA methylation changes (4951). Obesity, as measured by BMI, is also associated with distinct DNA methylation patterns (25, 28, 52, 53). In the current study, we did not find any associations between global methylation and BMI, habitual physical activity, or diet (total energy) in any of the cell subsets. These data do not support a role of adiposity, physical activity, or caloric intake on the altered global DNA methylation in PBMCs from women with PCOS. Interestingly, we found a negative association between global methylation in T helper cells and circulating levels of AMH. This relationship is only present in women with PCOS who have elevated circulating levels of AMH, suggesting a mechanistic association of this hormone in PCOS and DNA methylation changes in T helper cells. AMH is a member of the TGF-β ligand superfamily that is predominantly present in ovaries in healthy women but released into the circulation at elevated levels in PCOS (2, 5459), although there is little literature available indicating whether it is a cause or consequence. AMH has been found to cluster with many clinical markers of PCOS, including positive correlations with LH and androgens and negative correlations with FSH and glucose levels (58, 59). Furthermore, in women with PCOS, it has been shown that AMH disrupts folliculogenesis by decreasing sensitivity to FSH and thereby inhibiting follicle recruitment and growth, which can result in increased numbers of preantral and antral follicles and the PCO morphology (60, 61). It has also been suggested that circulating AMH may have a functional role outside of the reproductive system (62). Interestingly, in immune cells, members of the TGF-β ligand superfamily are potent regulators of T cell activation and differentiation and control a variety of regulatory epigenetic signals such as chromatin remodeling, histone modification, and DNA methylation (6365). Based on our observations and considering the literature, we hypothesize that AMH participates in the epigenetic reprogramming of some subpopulations of PBMCs in women with PCOS, warranting further research.

In this study, we identified a negative correlation between global methylation in T helper cells and fT. In women with PCOS, fT is elevated and is used to diagnose hyperandrogenism in the syndrome (31). Elevated androgens in animal models are associated with reproductive dysfunction (oligo-anovulation, menstrual disturbances, and subfertility) that is observed in PCOS (60, 6669). Androgens have immunomodulatory effects, and elevated androgens are associated with altered immune function that likely impacts reproductive function (70, 71). Medawar (72) identified the importance of the immune system in reproduction with further studies identifying the importance in the frequencies of T helper (Th)1, Th2, Th17, and regulatory T (Treg) cells in maintenance of normal ovarian function and menstrual cycles (7275). Interestingly, Th1/Th2/Th17 imbalances have been identified in women with PCOS (7577). The differentiation of T cells and the ratios of Th1/Th2/Th17/Treg cells appear to be modulated by androgens (71, 7880). Furthermore, the differentiation of T cells are also modulated by epigenetic mechanisms (15, 81, 82), and this may be the case in PCOS (71). Collectively, our data complements previous studies indicating that the elevated fT was associated with differences in the methylome profile in T helper cells in women with PCOS. Whether hyperandrogenism modulates the epigenome of T helper cells and result in changes to frequency of Th1/Th2/Th17/Treg cells and the reproductive dysfunction in PCOS would be an interesting avenue to explore.

DNA methylome and transcriptomic analysis of T helper cells

We also investigated genome-wide DNA methylation in T helper cells at the single nucleotide resolution and identified differentially methylated cytosine bases at the proximity of genes controlling reproductive function. Specifically, our gene ontology analysis returned terms such as female pregnancy, regulation of ovarian follicle development, and male sex determination.

As the altered DNA methylation was present at genes controlling reproductive function in immune cells, this suggests that epigenetic changes may not be tissue specific. Instead, these methylation patterns may occur in multiple tissue types that are exposed to specific extracellular stimuli (possibly elevated AMH levels or elevated androgens), with such exposure potentially occurring in utero or as a result of the development of PCOS during the lifespan (8385). With epigenetic reprogramming of genes being related to reproductive function, it would be worthwhile investigating other tissues, such as the ovary or neuroendocrine tissues, where the appropriate transcriptional activators and coactivators are expressed (86, 87). This would provide insights into the role of epigenetic reprogramming at genes controlling reproductive function on ovarian function and sex hormone imbalances in women with PCOS.

We found no overlap between the transcriptomic and DNA methylation data. This suggests that the shift in epigenetic signature could be related to differences in the proportions of T helper subpopulations (Th1, Th2, Th9, Th17, follicular T helper, regulatory, naive, effector, and memory T cells) (8890). Previous literature has shown in women with PCOS an increased frequency of the proinflammatory Th1 population and a reduced frequency of the anti-inflammatory Th2 population (75, 77, 78). Although we were unable to confirm the frequency of T helper subpopulations, the gene ontology pathway analysis identified differential methylation in the activation and proliferation of different T helper subpopulations, suggesting that women with PCOS may indeed have different proportions of the T helper subtypes (37).

To further consolidate this hypothesis, the transcriptomic analysis of the differentially expressed genes in T helper cells identified pathways that relate to T cell activation and differentiation. Of interest, KEGG analysis revealed that the BMP signaling pathway was upregulated in women with PCOS, and BMP, similar to AMH, is a member of the TGF-β ligand superfamily (91). The elevated circulating AMH found in our women with PCOS may explain the upregulation of BMP signaling in T helper cells via activation of the BMP receptors. AMH intracellular signaling works via the BMP receptor through the Smad1/5/8 pathway, which assembles into a complex with Smad4 and translocates into the nucleus, activating a range of genes dependent on the cellular context (9294). In T cells, BMP signaling generally regulates activation and differentiation of circulating naive immune cells, as well as proliferation of Th9, Th17, and memory T cells (94, 95). Despite the lack of quantification of T helper cell frequency in women with and without PCOS, our data provide a potential mechanism by which the upregulated BMP signaling pathway could impact the Th1/Th2 balance, but further studies are warranted (92, 96).

Several differentially expressed genes were associated with inflammation/inflammatory status (Table 5) in women with PCOS compared with those without. This supports the role of inflammation/inflammatory status consistent with previous studies (97), where ovulation requires an appropriate inflammatory reaction (18). Additionally, circulating immune cells and the cytokines they produce are also involved in ovarian function (16, 98, 99), suggesting that the epigenetic marks in our study can alter the transcriptome toward a proinflammatory T helper phenotype and suppress ovarian function.

Many of the differentially expressed genes identified from our transcriptomic analysis were noncoding, with most being lncRNAs, including pseudogenes, antisense RNA, and long intergenic noncoding RNAs. lncRNAs are widely expressed and regulate gene expression, particularly during development, differentiation, and activation of immune cells (100), and have been implicated in the comorbidities associated with PCOS, including T2DM (101, 102) and inflammatory disorders (103). There is also evidence to suggest that lncRNAs can act as a molecular scaffold for epigenetic modifications, including DNA methylation and histone modifications (104, 105). Indeed, the lack of overlap in our analysis between DNA methylation and gene expression suggests that other mechanisms, such as lncRNA, modulate the transcriptome in PCOS.

The strength of this study is that this cohort was a well-characterized, community-recruited group of women with and without PCOS, who were otherwise healthy. We were adequately powered to answer our a priori aims, with post hoc analysis revealing that we were powered on average 93% for all immune cell populations and had large effect sizes of ∼0.90 (106). We acknowledge that there is a need for more definitive research into the molecular mechanisms behind each of the different the phenotypes of PCOS. Although in the current study we were unable to confidently address the potential epigenetic differences between phenotypes in PCOS, we were able to explore the heterogeneous immune cell population and the unique role that these may play in PCOS. These findings provide novel avenues for future research in PCOS, including studying the role of methylation in specific immune cells in determining the different phenotypes in PCOS.

In conclusion, our study in a small, yet well-characterized cohort of women with and without PCOS demonstrates novel epigenomic insights into PCOS. The specific epigenetic reprogramming of genes involved in reproductive function in immune cells from women with PCOS is intriguing and may indicate a role for epigenetic factors in the reproductive dysfunction and sex hormone imbalance associated with PCOS. Finally, we found that global DNA methylation in T helper cells is negatively associated with circulating levels of AMH and fT, suggesting the importance of the cellular milieu (elevated circulating androgens and AMH) in the programming of T helper cells in PCOS and warranting further investigation.

Acknowledgments

We acknowledge Dr. Anju Joham and Dr. Soulmaz Shorakae for confirming diagnosis of women with PCOS, Prof. Helena Teede for critical review of the study design and expertise, and Dr. Sarah Voisin for assistance with statistical analysis.

Financial Support: This work was supported by the Novo Nordisk Foundation Center for Basic Metabolic Research, an independent research center at the University of Copenhagen partially funded by an unrestricted donation from the Novo Nordisk Foundation and by the National Health and Medical Research Council Centre for Research Excellence in PCOS. These funding bodies did not have a role in study design and have no role in the implementation of the study.

Additional Information

Disclosure Summary: The authors have nothing to disclose.

Data Availability: All data generated or analyzed during this study are included in this published article or in the data repositories listed in References.

Abbreviations:

    Abbreviations:
     
  • AMH

    anti-Müllerian hormone

  •  
  • BMI

    body mass index

  •  
  • BMP

    bone morphogenic protein

  •  
  • cGMP

    cyclic guanosine monophosphate

  •  
  • fT

    free testosterone

  •  
  • GO

    gene ontology

  •  
  • KEGG

    Kyoto Encyclopedia of Genes and Genomes

  •  
  • lncRNA

    long noncoding RNA

  •  
  • OGTT

    oral glucose tolerance test

  •  
  • PBMC

    peripheral blood mononuclear cell

  •  
  • PCOS

    polycystic ovary syndrome

  •  
  • RRBS

    reduced representation bisulfite sequencing

  •  
  • SAM

    S-adenosylmethionine

  •  
  • T2DM

    type 2 diabetes mellitus

  •  
  • Treg

    T regulatory

References and Notes

1.

Shorakae
S
,
Boyle
J
,
Teede
H
.
Polycystic ovary syndrome: a common hormonal condition with major metabolic sequelae that physicians should know about
.
Intern Med J
.
2014
;
44
(
8
):
720
726
.

2.

Cassar
S
,
Teede
HJ
,
Moran
LJ
,
Joham
AE
,
Harrison
CL
,
Strauss
BJ
,
Stepto
NK
.
Polycystic ovary syndrome and anti-Müllerian hormone: role of insulin resistance, androgens, obesity and gonadotrophins
.
Clin Endocrinol (Oxf)
.
2014
;
81
(
6
):
899
906
.

3.

Dumesic
DA
,
Oberfield
SE
,
Stener-Victorin
E
,
Marshall
JC
,
Laven
JS
,
Legro
RS
.
Scientific statement on the diagnostic criteria, epidemiology, pathophysiology, and molecular genetics of polycystic ovary syndrome
.
Endocr Rev
.
2015
;
36
(
5
):
487
525
.

4.

Legro
RS
,
Driscoll
D
,
Strauss
JF
III
,
Fox
J
,
Dunaif
A
.
Evidence for a genetic basis for hyperandrogenemia in polycystic ovary syndrome
.
Proc Natl Acad Sci USA
.
1998
;
95
(
25
):
14956
14960
.

5.

Legro
RS
,
Bentley-Lewis
R
,
Driscoll
D
,
Wang
SC
,
Dunaif
A
.
Insulin resistance in the sisters of women with polycystic ovary syndrome: association with hyperandrogenemia rather than menstrual irregularity
.
J Clin Endocrinol Metab
.
2002
;
87
(
5
):
2128
2133
.

6.

Day
FR
,
Hinds
DA
,
Tung
JY
,
Stolk
L
,
Styrkarsdottir
U
,
Saxena
R
,
Bjonnes
A
,
Broer
L
,
Dunger
DB
,
Halldorsson
BV
,
Lawlor
DA
,
Laval
G
,
Mathieson
I
,
McCardle
WL
,
Louwers
Y
,
Meun
C
,
Ring
S
,
Scott
RA
,
Sulem
P
,
Uitterlinden
AG
,
Wareham
NJ
,
Thorsteinsdottir
U
,
Welt
C
,
Stefansson
K
,
Laven
JSE
,
Ong
KK
,
Perry
JRB
.
Causal mechanisms and balancing selection inferred from genetic associations with polycystic ovary syndrome
.
Nat Commun
.
2015
;
6
(
1
):
8464
.

7.

Hayes
MG
,
Urbanek
M
,
Ehrmann
DA
,
Armstrong
LL
,
Lee
JY
,
Sisk
R
,
Karaderi
T
,
Barber
TM
,
McCarthy
MI
,
Franks
S
,
Lindgren
CM
,
Welt
CK
,
Diamanti-Kandarakis
E
,
Panidis
D
,
Goodarzi
MO
,
Azziz
R
,
Zhang
Y
,
James
RG
,
Olivier
M
,
Kissebah
AH
,
Stener-Victorin
E
,
Legro
RS
,
Dunaif
A
;
Reproductive Medicine Network
.
Genome-wide association of polycystic ovary syndrome implicates alterations in gonadotropin secretion in European ancestry populations [published correction appears in Nat Commun. 2016;7:10762]
.
Nat Commun
.
2015
;
6
(
1
):
7502
.

8.

Shi
Y
,
Zhao
H
,
Shi
Y
,
Cao
Y
,
Yang
D
,
Li
Z
,
Zhang
B
,
Liang
X
,
Li
T
,
Chen
J
,
Shen
J
,
Zhao
J
,
You
L
,
Gao
X
,
Zhu
D
,
Zhao
X
,
Yan
Y
,
Qin
Y
,
Li
W
,
Yan
J
,
Wang
Q
,
Zhao
J
,
Geng
L
,
Ma
J
,
Zhao
Y
,
He
G
,
Zhang
A
,
Zou
S
,
Yang
A
,
Liu
J
,
Li
W
,
Li
B
,
Wan
C
,
Qin
Y
,
Shi
J
,
Yang
J
,
Jiang
H
,
Xu
JE
,
Qi
X
,
Sun
Y
,
Zhang
Y
,
Hao
C
,
Ju
X
,
Zhao
D
,
Ren
CE
,
Li
X
,
Zhang
W
,
Zhang
Y
,
Zhang
J
,
Wu
D
,
Zhang
C
,
He
L
,
Chen
ZJ
.
Genome-wide association study identifies eight new risk loci for polycystic ovary syndrome
.
Nat Genet
.
2012
;
44
(
9
):
1020
1025
.

9.

Chen
ZJ
,
Zhao
H
,
He
L
,
Shi
Y
,
Qin
Y
,
Shi
Y
,
Li
Z
,
You
L
,
Zhao
J
,
Liu
J
,
Liang
X
,
Zhao
X
,
Zhao
J
,
Sun
Y
,
Zhang
B
,
Jiang
H
,
Zhao
D
,
Bian
Y
,
Gao
X
,
Geng
L
,
Li
Y
,
Zhu
D
,
Sun
X
,
Xu
JE
,
Hao
C
,
Ren
CE
,
Zhang
Y
,
Chen
S
,
Zhang
W
,
Yang
A
,
Yan
J
,
Li
Y
,
Ma
J
,
Zhao
Y
.
Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3
.
Nat Genet
.
2011
;
43
(
1
):
55
59
.

10.

Shen
HR
,
Qiu
LH
,
Zhang
ZQ
,
Qin
YY
,
Cao
C
,
Di
W
.
Genome-wide methylated DNA immunoprecipitation analysis of patients with polycystic ovary syndrome
.
PLoS One
.
2013
;
8
(
5
):
e64801
.

11.

Wang
XX
,
Wei
JZ
,
Jiao
J
,
Jiang
SY
,
Yu
DH
,
Li
D
.
Genome-wide DNA methylation and gene expression patterns provide insight into polycystic ovary syndrome development
.
Oncotarget
.
2014
;
5
(
16
):
6603
6610
.

12.

Li
S
,
Zhu
D
,
Duan
H
,
Ren
A
,
Glintborg
D
,
Andersen
M5
,
Skov
V
,
Thomassen
M
,
Kruse
T
,
Tan
Q
.
Differential DNA methylation patterns of polycystic ovarian syndrome in whole blood of Chinese women
.
Oncotarget
. Oncotarget.
2017
;
8
(
13
):
20656
20666
.

13.

Kokosar
M
,
Benrick
A
,
Perfilyev
A
,
Fornes
R
,
Nilsson
E
,
Maliqueo
M
,
Behre
CJ
,
Sazonova
A
,
Ohlsson
C
,
Ling
C
,
Stener-Victorin
E
.
Epigenetic and transcriptional alterations in human adipose tissue of polycystic ovary syndrome [published correction appears in Sci Rep. 2016;6:25321]
.
Sci Rep
.
2016
;
6
(
1
):
22883
.

14.

Nilsson
E
,
Benrick
A
,
Kokosar
M
,
Krook
A
,
Lindgren
E
,
Källman
T
,
Martis
MM
,
Højlund
K
,
Ling
C
,
Stener-Victorin
E
.
Transcriptional and epigenetic changes influencing skeletal muscle metabolism in women with polycystic ovary syndrome
.
J Clin Endocrinol Metab
.
2018
;
103
(
12
):
4465
4477
.

15.

Raghuraman
S
,
Donkin
I
,
Versteyhe
S
,
Barrès
R
,
Simar
D
.
The emerging role of epigenetics in inflammation and immunometabolism
.
Trends Endocrinol Metab
.
2016
;
27
(
11
):
782
795
.

16.

Clancy
KB
,
Baerwald
AR
,
Pierson
RA
.
Systemic inflammation is associated with ovarian follicular dynamics during the human menstrual cycle
.
PLoS One
.
2013
;
8
(
5
):
e64807
.

17.

Brännström
M
,
Norman
RJ
.
Involvement of leukocytes and cytokines in the ovulatory process and corpus luteum function
.
Hum Reprod
.
1993
;
8
(
10
):
1762
1775
.

18.

Espey
LL
.
Ovulation as an inflammatory reaction—a hypothesis
.
Biol Reprod
.
1980
;
22
(
1
):
73
106
.

19.

Bukovsky
A
,
Presl
J
.
Ovarian function and the immune system
.
Med Hypotheses
.
1979
;
5
(
4
):
415
436
.

20.

Pate
JL
,
Toyokawa
K
,
Walusimbi
S
,
Brzezicka
E
.
The interface of the immune and reproductive systems in the ovary: lessons learned from the corpus luteum of domestic animal models
.
Am J Reprod Immunol
.
2010
;
64
(
4
):
275
286
.

21.

Figueroa
F
,
Davicino
R
,
Micalizzi
B
,
Oliveros
L
,
Forneris
M
.
Macrophage secretions modulate the steroidogenesis of polycystic ovary in rats: effect of testosterone on macrophage pro-inflammatory cytokines
.
Life Sci
.
2012
;
90
(
19–20
):
733
739
.

22.

Schmidt
J
,
Weijdegård
B
,
Mikkelsen
AL
,
Lindenberg
S
,
Nilsson
L
,
Brännström
M
.
Differential expression of inflammation-related genes in the ovarian stroma and granulosa cells of PCOS women
.
Mol Hum Reprod
.
2014
;
20
(
1
):
49
58
.

23.

Zierath
JR
,
Barrès
RE
.
Nutritional status affects the epigenomic profile of peripheral blood cells
.
Epigenomics
.
2011
;
3
(
3
):
259
260
.

24.

Delamaire
M
,
Maugendre
D
,
Moreno
M
,
Le Goff
MC
,
Allannic
H
,
Genetet
B
.
Impaired leucocyte functions in diabetic patients
.
Diabet Med
.
1997
;
14
(
1
):
29
34
.

25.

Wang
X
,
Zhu
H
,
Snieder
H
,
Su
S
,
Munn
D
,
Harshfield
G
,
Maria
BL
,
Dong
Y
,
Treiber
F
,
Gutin
B
,
Shi
H
.
Obesity related methylation changes in DNA of peripheral blood leukocytes
.
BMC Med
.
2010
;
8
(
1
):
87
.

26.

Kintscher
U
,
Hartge
M
,
Hess
K
,
Foryst-Ludwig
A
,
Clemenz
M
,
Wabitsch
M
,
Fischer-Posovszky
P
,
Barth
TF
,
Dragun
D
,
Skurk
T
,
Hauner
H
,
Blüher
M
,
Unger
T
,
Wolf
AM
,
Knippschild
U
,
Hombach
V
,
Marx
N
.
T-lymphocyte infiltration in visceral adipose tissue: a primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance
.
Arterioscler Thromb Vasc Biol
.
2008
;
28
(
7
):
1304
1310
.

27.

Xu
N
,
Azziz
R
,
Goodarzi
MO
.
Epigenetics in polycystic ovary syndrome: a pilot study of global DNA methylation
.
Fertil Steril
.
2010
;
94
(
2
):
781
783.e1
.

28.

Simar
D
,
Versteyhe
S
,
Donkin
I
,
Liu
J
,
Hesson
L
,
Nylander
V
,
Fossum
A
,
Barrès
R
.
DNA methylation is altered in B and NK lymphocytes in obese and type 2 diabetic human
.
Metabolism
.
2014
;
63
(
9
):
1188
1197
.

29.

Adalsteinsson
BT
,
Gudnason
H
,
Aspelund
T
,
Harris
TB
,
Launer
LJ
,
Eiriksdottir
G
,
Smith
AV
,
Gudnason
V
.
Heterogeneity in white blood cells has potential to confound DNA methylation measurements
.
PLoS One
.
2012
;
7
(
10
):
e46705
.

30.

Glossop
JR
,
Nixon
NB
,
Emes
RD
,
Haworth
KE
,
Packham
JC
,
Dawes
PT
,
Fryer
AA
,
Mattey
DL
,
Farrell
WE
.
Epigenome-wide profiling identifies significant differences in DNA methylation between matched-pairs of T- and B-lymphocytes from healthy individuals
.
Epigenetics
.
2013
;
8
(
11
):
1188
1197
.

31.

Rotterdam ESHRE/ASRM-Sponsored PCOS Consensus Workshop Group
.
Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome
.
Fertil Steril
.
2004
;
81
(
1
):
19
25
.

32.

Teede
HJ
,
Misso
ML
,
Costello
MF
,
Dokras
A
,
Laven
J
,
Moran
L
,
Piltonen
T
,
Norman
RJ
;
International PCOS Network
.
Recommendations from the international evidence-based guideline for the assessment and management of polycystic ovary syndrome
.
Fertil Steril
.
2018
;
110
(
3
):
364
379
.

33.

Trost
SG
,
Pate
RR
,
Sallis
JF
,
Freedson
PS
,
Taylor
WC
,
Dowda
M
,
Sirard
J
.
Age and gender differences in objectively measured physical activity in youth
.
Med Sci Sports Exerc
.
2002
;
34
(
2
):
350
355
.

34.

American College of Sports Medicine
;
Swain
DP
, ed.
ACSM’s Resource Manual for Guidelines for Exercise Testing and Prescription
. 7th ed.
Philadelphia, PA
:
Wolters Kluwer Health/Lippincott Williams & Wilkins
;
2014
.

35.

Meyer
C
,
McGrath
BP
,
Teede
HJ
.
Effects of medical therapy on insulin resistance and the cardiovascular system in polycystic ovary syndrome
.
Diabetes Care
.
2007
;
30
(
3
):
471
478
.

36.

Södergård
R
,
Bäckström
T
,
Shanbhag
V
,
Carstensen
H
.
Calculation of free and bound fractions of testosterone and estradiol-17β to human plasma proteins at body temperature
.
J Steroid Biochem
.
1982
;
16
(
6
):
801
810
.

37.

Hiam
D
,
Simar
D
,
Laker
R
,
Altıntaş
A
,
Gibson-Helm
M
,
Fletcher
E
,
Moreno-Asso
A
,
Trewin
AJ
,
Barres
R
,
Stepto
NK
.
Data from: Epigenetic reprogramming of immune cells in women with PCOS impact genes controlling reproductive function. figshare 2019. Deposited 5 January 2019. https://figshare.com/s/8088ab38eb90d059dc86
.

38.

Laker
RC
,
Garde
C
,
Camera
DM
,
Smiles
WJ
,
Zierath
JR
,
Hawley
JA
,
Barrès
R
.
Transcriptomic and epigenetic responses to short-term nutrient-exercise stress in humans [published correction appears in Sci Rep. 2018;8(1):5008]
.
Sci Rep
.
2017
;
7
(
1
):
15134
.

39.

Dobin
A
,
Davis
CA
,
Schlesinger
F
,
Drenkow
J
,
Zaleski
C
,
Jha
S
,
Batut
P
,
Chaisson
M
,
Gingeras
TR
.
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
.
2013
;
29
(
1
):
15
21
.

40.

Liao
Y
,
Smyth
GK
,
Shi
W
.
featureCounts: an efficient general purpose program for assigning sequence reads to genomic features
.
Bioinformatics
.
2014
;
30
(
7
):
923
930
.

41.

Harrow
J
,
Frankish
A
,
Gonzalez
JM
,
Tapanari
E
,
Diekhans
M
,
Kokocinski
F
,
Aken
BL
,
Barrell
D
,
Zadissa
A
,
Searle
S
,
Barnes
I
,
Bignell
A
,
Boychenko
V
,
Hunt
T
,
Kay
M
,
Mukherjee
G
,
Rajan
J
,
Despacio-Reyes
G
,
Saunders
G
,
Steward
C
,
Harte
R
,
Lin
M
,
Howald
C
,
Tanzer
A
,
Derrien
T
,
Chrast
J
,
Walters
N
,
Balasubramanian
S
,
Pei
B
,
Tress
M
,
Rodriguez
JM
,
Ezkurdia
I
,
van Baren
J
,
Brent
M
,
Haussler
D
,
Kellis
M
,
Valencia
A
,
Reymond
A
,
Gerstein
M
,
Guigó
R
,
Hubbard
TJ
.
GENCODE: the reference human genome annotation for the ENCODE Project
.
Genome Res
.
2012
;
22
(
9
):
1760
1774
.

42.

Love
MI
,
Huber
W
,
Anders
S
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
.
2014
;
15
(
12
):
550
.

43.

Krueger
F
,
Andrews
SR
.
Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications
.
Bioinformatics
.
2011
;
27
(
11
):
1571
1572
.

44.

Assenov
Y
,
Müller
F
,
Lutsik
P
,
Walter
J
,
Lengauer
T
,
Bock
C
.
Comprehensive analysis of DNA methylation data with RnBeads
.
Nat Methods
.
2014
;
11
(
11
):
1138
1140
.

45.

Hiam
D
,
Simar
D
,
Laker
R
,
Altıntaş
A
,
Gibson-Helm
M
,
Fletcher
E
,
Moreno-Asso
A
,
Trewin
AJ
,
Barres
R
,
Stepto
NK
.
Data from: Epigenetic reprogramming of immune cells in women with PCOS impact genes controlling reproductive function. NCBI GEO. Deposited 5 April 2019. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE130582
.

46.

Rotterdam ESHRE/ASRM-Sponsored PCOS consensus workshop group
.
Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS)
.
Hum Reprod
.
2004
;
19
(
1
):
41
47
.

47.

Crider
KS
,
Yang
TP
,
Berry
RJ
,
Bailey
LB
.
Folate and DNA methylation: a review of molecular mechanisms and the evidence for folate’s role
.
Adv Nutr
.
2012
;
3
(
1
):
21
38
.

48.

Tajima
S
,
Suetake
I
,
Takeshita
K
,
Nakagawa
A
,
Kimura
H
.
Domain structure of the Dnmt1, Dnmt3a, and Dnmt3b DNA methyltransferases
.
Adv Exp Med Biol
.
2016
;
945
:
63
86
.

49.

Feil
R
,
Fraga
MF
.
Epigenetics and the environment: emerging patterns and implications
.
Nat Rev Genet
.
2012
;
13
(
2
):
97
109
.

50.

Mendelson
MM
,
Marioni
RE
,
Joehanes
R
,
Liu
C
,
Hedman
ÅK
,
Aslibekyan
S
,
Demerath
EW
,
Guan
W
,
Zhi
D
,
Yao
C
,
Huan
T
,
Willinger
C
,
Chen
B
,
Courchesne
P
,
Multhaup
M
,
Irvin
MR
,
Cohain
A
,
Schadt
EE
,
Grove
ML
,
Bressler
J
,
North
K
,
Sundström
J
,
Gustafsson
S
,
Shah
S
,
McRae
AF
,
Harris
SE
,
Gibson
J
,
Redmond
P
,
Corley
J
,
Murphy
L
,
Starr
JM
,
Kleinbrink
E
,
Lipovich
L
,
Visscher
PM
,
Wray
NR
,
Krauss
RM
,
Fallin
D
,
Feinberg
A
,
Absher
DM
,
Fornage
M
,
Pankow
JS
,
Lind
L
,
Fox
C
,
Ingelsson
E
,
Arnett
DK
,
Boerwinkle
E
,
Liang
L
,
Levy
D
,
Deary
IJ
.
Association of body mass index with DNA methylation and gene expression in blood cells and relations to cardiometabolic disease: a Mendelian randomization approach
.
PLoS Med
.
2017
;
14
(
1
):
e1002215
.

51.

Rasmussen
M
,
Zierath
JR
,
Barrès
R
.
Dynamic epigenetic responses to muscle contraction
.
Drug Discov Today
.
2014
;
19
(
7
):
1010
1014
.

52.

Barres
R
,
Kirchner
H
,
Rasmussen
M
,
Yan
J
,
Kantor
FR
,
Krook
A
,
Näslund
E
,
Zierath
JR
.
Weight loss after gastric bypass surgery in human obesity remodels promoter methylation [published correction appears in Cell Rep. 2013;3(5):1755]
.
Cell Reports
.
2013
;
3
(
4
):
1020
1027
.

53.

Jacobsen
MJ
,
Mentzel
CM
,
Olesen
AS
,
Huby
T
,
Jørgensen
CB
,
Barrès
R
,
Fredholm
M
,
Simar
D
.
Altered methylation profile of lymphocytes is concordant with perturbation of lipids metabolism and inflammatory response in obesity
.
J Diabetes Res
.
2016
;
2016
:
8539057
.

54.

Teede
H
,
Misso
M
,
Tassone
EC
,
Dewailly
D
,
Ng
EH
,
Azziz
R
,
Norman
RJ
,
Andersen
M
,
Franks
S
,
Hoeger
K
,
Hutchison
S
,
Oberfield
S
,
Shah
D
,
Hohmann
F
,
Ottey
S
,
Dabadghao
P
,
Laven
JSE
.
Anti-Müllerian hormone in PCOS: a review informing international guidelines
.
Trends Endocrinol Metab
.
2019
;
30
(
7
):
467
478
.

55.

Cook
CL
,
Siow
Y
,
Brenner
AG
,
Fallat
ME
.
Relationship between serum müllerian-inhibiting substance and other reproductive hormones in untreated women with polycystic ovary syndrome and normal women
.
Fertil Steril
.
2002
;
77
(
1
):
141
146
.

56.

Siow
Y
,
Kives
S
,
Hertweck
P
,
Perlman
S
,
Fallat
ME
.
Serum Müllerian-inhibiting substance levels in adolescent girls with normal menstrual cycles or with polycystic ovary syndrome
.
Fertil Steril
.
2005
;
84
(
4
):
938
944
.

57.

Lie Fong
S
,
Laven
JSE
,
Duhamel
A
,
Dewailly
D
.
Polycystic ovarian morphology and the diagnosis of polycystic ovary syndrome: redefining threshold levels for follicle count and serum anti-Müllerian hormone using cluster analysis
.
Hum Reprod
.
2017
;
32
(
8
):
1723
1731
.

58.

Sova
H
,
Unkila-Kallio
L
,
Tiitinen
A
,
Hippeläinen
M
,
Perheentupa
A
,
Tinkanen
H
,
Puukka
K
,
Bloigu
R
,
Piltonen
T
,
Tapanainen
JS
,
Morin-Papunen
L
.
Hormone profiling, including anti-Müllerian hormone (AMH), for the diagnosis of polycystic ovary syndrome (PCOS) and characterization of PCOS phenotypes
.
Gynecol Endocrinol
.
2019
;
35
(
7
):
595
600
.

59.

Tata
B
,
Mimouni
NEH
,
Barbotin
AL
,
Malone
SA
,
Loyens
A
,
Pigny
P
,
Dewailly
D
,
Catteau-Jonard
S
,
Sundström-Poromaa
I
,
Piltonen
TT
,
Dal Bello
F
,
Medana
C
,
Prevot
V
,
Clasadonte
J
,
Giacobini
P
.
Elevated prenatal anti-Müllerian hormone reprograms the fetus and induces polycystic ovary syndrome in adulthood
.
Nat Med
.
2018
;
24
(
6
):
834
846
.

60.

Lebbe
M
,
Woodruff
TK
.
Involvement of androgens in ovarian health and disease
.
Mol Hum Reprod
.
2013
;
19
(
12
):
828
837
.

61.

Catteau-Jonard
S
,
Jamin
SP
,
Leclerc
A
,
Gonzalès
J
,
Dewailly
D
,
di Clemente
N
.
Anti-Mullerian hormone, its receptor, FSH receptor, and androgen receptor genes are overexpressed by granulosa cells from stimulated follicles in women with polycystic ovary syndrome
.
J Clin Endocrinol Metab
.
2008
;
93
(
11
):
4456
4461
.

62.

Stepto
NK
,
Moreno-Asso
A
,
McIlvenna
LC
,
Walters
KA
,
Rodgers
RJ
.
Molecular mechanisms of insulin resistance in polycystic ovary syndrome. Unraveling the conundrum in skeletal muscle
?
J Clin Endocrinol Metab
.
2019
;
104
(
11
):
5372
5381
.

63.

Bai
J
,
Xi
Q
.
Crosstalk between TGF-β signaling and epigenome
.
Acta Biochim Biophys Sin (Shanghai)
.
2018
;
50
(
1
):
60
67
.

64.

Konkel
JE
,
Zhang
D
,
Zanvit
P
,
Chia
C
,
Zangarle-Murray
T
,
Jin
W
,
Wang
S
,
Chen
W
.
Transforming growth factor-β signaling in regulatory T cells controls T helper-17 cells and tissue-specific immune responses
.
Immunity
.
2017
;
46
(
4
):
660
674
.

65.

Li
MO
,
Wan
YY
,
Sanjabi
S
,
Robertson
A-KL
,
Flavell
RA
.
Transforming growth factor-β regulation of immune responses
.
Annu Rev Immunol
.
2006
;
24
(
1
):
99
146
.

66.

Teede
H
,
Deeks
A
,
Moran
L
.
Polycystic ovary syndrome: a complex condition with psychological, reproductive and metabolic manifestations that impacts on health across the lifespan
.
BMC Med
.
2010
;
8
(
1
):
41
.

67.

Bishop
CV
,
Reiter
TE
,
Erikson
DW
,
Hanna
CB
,
Daughtry
BL
,
Chavez
SL
,
Hennebold
JD
,
Stouffer
RL
.
Chronically elevated androgen and/or consumption of a Western-style diet impairs oocyte quality and granulosa cell function in the nonhuman primate periovulatory follicle
.
J Assist Reprod Genet
.
2019
;
36
(
7
):
1497
1511
.

68.

Catteau-Jonard
S
,
Dewailly
D
.
Pathophysiology of polycystic ovary syndrome: the role of hyperandrogenism
.
Front Horm Res
.
2013
;
40
:
22
27
.

69.

Jonard
S
,
Robert
Y
,
Cortet-Rudelli
C
,
Pigny
P
,
Decanter
C
,
Dewailly
D
.
Ultrasound examination of polycystic ovaries: is it worth counting the follicles
?
Hum Reprod
.
2003
;
18
(
3
):
598
603
.

70.

Segner
H
,
Verburg-van Kemenade
BML
,
Chadzinska
M
.
The immunomodulatory role of the hypothalamus-pituitary-gonad axis: proximate mechanism for reproduction-immune trade offs
?
Dev Comp Immunol
.
2017
;
66
:
43
60
.

71.

Walecki
M
,
Eisel
F
,
Klug
J
,
Baal
N
,
Paradowska-Dogan
A
,
Wahle
E
,
Hackstein
H
,
Meinhardt
A
,
Fijak
M
.
Androgen receptor modulates Foxp3 expression in CD4+CD25+Foxp3+ regulatory T-cells
.
Mol Biol Cell
.
2015
;
26
(
15
):
2845
2857
.

72.

Medawar
P
.
Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates
.
Symp Soc Exp Biol
.
1953
;
7
:
320
338
.

73.

Polese
B
,
Gridelet
V
,
Araklioti
E
,
Martens
H
,
Perrier d’Hauterive
S
,
Geenen
V
.
The endocrine milieu and CD4 T-lymphocyte polarization during pregnancy
.
front endocrinol (lausanne)
.
2014
;
5
:
106
.

74.

Abrams
ET
,
Miller
EM
.
The roles of the immune system in women’s reproduction: evolutionary constraints and life history trade-offs
.
Am J Phys Anthropol
.
2011
;
146
(
Suppl 53
):
134
154
.

75.

Omu
AE
,
Al-Azemi
MK
,
Makhseed
M
,
Al-Oattan
F
,
Ismail
AA
,
Al-Tahir
S
,
Al-Busiri
N
.
Differential expression of T-helper cytokines in the peritoneal fluid of women with normal ovarian cycle compared with women with chronic anovulation
.
Acta Obstet Gynecol Scand
.
2003
;
82
(
7
):
603
609
.

76.

Nasri
F
,
Doroudchi
M
,
Namavar Jahromi
B
,
Gharesi-Fard
B
.
T helper cells profile and CD4+CD25+Foxp3+ regulatory T cells in polycystic ovary syndrome
.
Iran J Immunol
.
2018
;
15
(
3
):
175
185
.

77.

Qin
L
,
Xu
W
,
Li
X
,
Meng
W
,
Hu
L
,
Luo
Z
,
Wang
Y
,
Luo
S
,
Li
S
.
Differential expression profile of immunological cytokines in local ovary in patients with polycystic ovarian syndrome: analysis by flow cytometry
.
Eur J Obstet Gynecol Reprod Biol
.
2016
;
197
:
136
141
.

78.

Gong
P
,
Shi
B
,
Wang
J
,
Cao
P
,
Diao
Z
,
Wang
Y
,
Hu
Y
,
Li
S
.
Association between Th1/Th2 immune imbalance and obesity in women with or without polycystic ovary syndrome
.
Gynecol Endocrinol
.
2018
;
34
(
8
):
709
714
.

79.

Olsen
NJ
,
Kovacs
WJ
.
Effects of androgens on T and B lymphocyte development
.
Immunol Res
.
2001
;
23
(
2-3
):
281
288
.

80.

Trigunaite
A
,
Dimo
J
,
Jørgensen
TN
.
Suppressive effects of androgens on the immune system
.
Cell Immunol
.
2015
;
294
(
2
):
87
94
.

81.

Floess
S
,
Freyer
J
,
Siewert
C
,
Baron
U
,
Olek
S
,
Polansky
J
,
Schlawe
K
,
Chang
HD
,
Bopp
T
,
Schmitt
E
,
Klein-Hessling
S
,
Serfling
E
,
Hamann
A
,
Huehn
J
.
Epigenetic control of the foxp3 locus in regulatory T cells
.
PLoS Biol
.
2007
;
5
(
2
):
e38
.

82.

Lee
DU
,
Agarwal
S
,
Rao
A
.
Th2 lineage commitment and efficient IL-4 production involves extended demethylation of the IL-4 gene
.
Immunity
.
2002
;
16
(
5
):
649
660
.

83.

Barry
JA
,
Kay
AR
,
Navaratnarajah
R
,
Iqbal
S
,
Bamfo
JE
,
David
AL
,
Hines
M
,
Hardiman
PJ
.
Umbilical vein testosterone in female infants born to mothers with polycystic ovary syndrome is elevated to male levels
.
J Obstet Gynaecol
.
2010
;
30
(
5
):
444
446
.

84.

Jennewein
MF
,
Abu-Raya
B
,
Jiang
Y
,
Alter
G
,
Marchant
A
.
Transfer of maternal immunity and programming of the newborn immune system
.
Semin Immunopathol
.
2017
;
39
(
6
):
605
613
.

85.

Hart
R
,
Sloboda
DM
,
Doherty
DA
,
Norman
RJ
,
Atkinson
HC
,
Newnham
JP
,
Dickinson
JE
,
Hickey
M
.
Circulating maternal testosterone concentrations at 18 weeks of gestation predict circulating levels of antimüllerian hormone in adolescence: a prospective cohort study
.
Fertil Steril
.
2010
;
94
(
4
):
1544
1547
.

86.

Fabre
O
,
Ingerslev
LR
,
Garde
C
,
Donkin
I
,
Simar
D
,
Barrès
R
.
Exercise training alters the genomic response to acute exercise in human adipose tissue
.
Epigenomics
.
2018
;
10
(
8
):
1033
��
1050
.

87.

Donkin
I
,
Barrès
R
.
Sperm epigenetics and influence of environmental factors
.
Mol Metab
.
2018
;
14
:
1
11
.

88.

Sellars
M
,
Huh
JR
,
Day
K
,
Issuree
PD
,
Galan
C
,
Gobeil
S
,
Absher
D
,
Green
MR
,
Littman
DR
.
Regulation of DNA methylation dictates Cd4 expression during the development of helper and cytotoxic T cell lineages
.
Nat Immunol
.
2015
;
16
(
7
):
746
754
.

89.

Li
Y
,
Chen
G
,
Ma
L
,
Ohms
SJ
,
Sun
C
,
Shannon
MF
,
Fan
JY
.
Plasticity of DNA methylation in mouse T cell activation and differentiation
.
BMC Mol Biol
.
2012
;
13
(
1
):
16
.

90.

Qiu
H
,
Wu
H
,
Chan
V
,
Lau
CS
,
Lu
Q
.
Transcriptional and epigenetic regulation of follicular T-helper cells and their role in autoimmunity
.
Autoimmunity
.
2017
;
50
(
2
):
71
81
.

91.

Akhurst
RJ
,
Hata
A
.
Targeting the TGFβ signalling pathway in disease [published correction appears in Nat Rev Drug Discov. 2012;11:886]
.
Nat Rev Drug Discov
.
2012
;
11
(
10
):
790
811
.

92.

Josso
N
,
Clemente
N
.
Transduction pathway of anti-Müllerian hormone, a sex-specific member of the TGF-β family
.
Trends Endocrinol Metab
.
2003
;
14
(
2
):
91
97
.

93.

Nohe
A
,
Hassel
S
,
Ehrlich
M
,
Neubauer
F
,
Sebald
W
,
Henis
YI
,
Knaus
P
.
The mode of bone morphogenetic protein (BMP) receptor oligomerization determines different BMP-2 signaling pathways
.
J Biol Chem
.
2002
;
277
(
7
):
5330
5338
.

94.

Martínez
VG
,
Sacedón
R
,
Hidalgo
L
,
Valencia
J
,
Fernández-Sevilla
LM
,
Hernández-López
C
,
Vicente
A
,
Varas
A
.
The BMP pathway participates in human naive CD4+ T cell activation and homeostasis
.
PLoS One
.
2015
;
10
(
6
):
e0131453
.

95.

Veldhoen
M
,
Uyttenhove
C
,
van Snick
J
,
Helmby
H
,
Westendorf
A
,
Buer
J
,
Martin
B
,
Wilhelm
C
,
Stockinger
B
.
Transforming growth factor-β “reprograms” the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset
.
Nat Immunol
.
2008
;
9
(
12
):
1341
1346
.

96.

Yoshioka
Y
,
Ono
M
,
Osaki
M
,
Konishi
I
,
Sakaguchi
S
.
Differential effects of inhibition of bone morphogenic protein (BMP) signalling on T-cell activation and differentiation
.
Eur J Immunol
.
2012
;
42
(
3
):
749
759
.

97.

Su
NJ
,
Ma
J
,
Feng
DF
,
Zhou
S
,
Li
ZT
,
Zhou
WP
,
Deng
H
,
Liang
JY
,
Yang
XH
,
Zhang
YM
,
Liu
FH
,
Zhang
L
.
The peripheral blood transcriptome identifies dysregulation of inflammatory response genes in polycystic ovary syndrome
.
Gynecol Endocrinol
.
2018
;
34
(
7
):
584
588
.

98.

Clancy
KB
,
Klein
LD
,
Ziomkiewicz
A
,
Nenko
I
,
Jasienska
G
,
Bribiescas
RG
.
Relationships between biomarkers of inflammation, ovarian steroids, and age at menarche in a rural Polish sample
.
Am J Hum Biol
.
2013
;
25
(
3
):
389
398
.

99.

Clancy
KB
,
Baerwald
AR
,
Pierson
RA
.
Cycle-phase dependent associations between CRP, leptin, and reproductive hormones in an urban, Canadian sample
.
Am J Phys Anthropol
.
2016
;
160
(
3
):
389
396
.

100.

Atianand
MK
,
Fitzgerald
KA
.
Long non-coding RNAs and control of gene expression in the immune system
.
Trends Mol Med
.
2014
;
20
(
11
):
623
631
.

101.

Kornfeld
JW
,
Brüning
JC
.
Regulation of metabolism by long, non-coding RNAs
.
Front Genet
.
2014
;
5
:
57
.

102.

Knoll
M
,
Lodish
HF
,
Sun
L
.
Long non-coding RNAs as regulators of the endocrine system
.
Nat Rev Endocrinol
.
2015
;
11
(
3
):
151
160
.

103.

Carpenter
S
,
Aiello
D
,
Atianand
MK
,
Ricci
EP
,
Gandhi
P
,
Hall
LL
,
Byron
M
,
Monks
B
,
Henry-Bezy
M
,
Lawrence
JB
,
O’Neill
LA
,
Moore
MJ
,
Caffrey
DR
,
Fitzgerald
KA
.
A long noncoding RNA mediates both activation and repression of immune response genes
.
Science
.
2013
;
341
(
6147
):
789
792
.

104.

Spitale
RC
,
Tsai
MC
,
Chang
HY
.
RNA templating the epigenome: long noncoding RNAs as molecular scaffolds
.
Epigenetics
.
2011
;
6
(
5
):
539
543
.

105.

Rinn
JL
,
Chang
HY
.
Genome regulation by long noncoding RNAs
.
Annu Rev Biochem
.
2012
;
81
(
1
):
145
166
.

106.

Faul
F
,
Erdfelder
E
,
Lang
AG
,
Buchner
A
.
G*Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences
.
Behav Res Methods
.
2007
;
39
(
2
):
175
191
.

Author notes

R.B. and N.K.S. are joint senior authors.