#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Androgens regulate ovarian gene expression by balancing Ezh2-Jmjd3 mediated H3K27me3 dynamics


Authors: Sambit Roy aff001;  Binbin Huang aff002;  Niharika Sinha aff001;  Jianrong Wang aff002;  Aritro Sen aff001
Authors place of work: Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, MI, United States of America aff001;  Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States of America aff002
Published in the journal: Androgens regulate ovarian gene expression by balancing Ezh2-Jmjd3 mediated H3K27me3 dynamics. PLoS Genet 17(3): e1009483. doi:10.1371/journal.pgen.1009483
Category: Research Article
doi: https://doi.org/10.1371/journal.pgen.1009483

Summary

Conventionally viewed as male hormone, androgens play a critical role in female fertility. Although androgen receptors (AR) are transcription factors, to date very few direct transcriptional targets of ARs have been identified in the ovary. Using mouse models, this study provides three critical insights about androgen-induced gene regulation in the ovary and its impact on female fertility. First, RNA-sequencing reveals a number of genes and biological processes that were previously not known to be directly regulated by androgens in the ovary. Second, androgens can also influence gene expression by decreasing the tri-methyl mark on lysine 27 of histone3 (H3K27me3), a gene silencing epigenetic mark. ChIP-seq analyses highlight that androgen-induced modulation of H3K27me3 mark within gene bodies, promoters or distal enhancers have a much broader impact on ovarian function than the direct genomic effects of androgens. Third, androgen-induced decrease of H3K27me3 is mediated through (a) inhibiting the expression and activity of Enhancer of Zeste Homologue 2 (EZH2), a histone methyltransferase that promotes tri-methylation of K27 and (b) by inducing the expression of a histone demethylase called Jumonji domain containing protein-3 (JMJD3/KDM6B), responsible for removing the H3K27me3 mark. Androgens through the PI3K/Akt pathway, in a transcription-independent fashion, increase hypoxia-inducible factor 1 alpha (HIF1α) protein levels, which in turn induce JMJD3 expression. Furthermore, proof of concept studies involving in vivo knockdown of Ar in the ovary and ovarian (granulosa) cell-specific Ar knockout mouse model show that ARs regulate the expression of key ovarian genes through modulation of H3K27me3.

Keywords:

Androgens – Gene expression – Gene regulation – Ovaries – Polycystic ovary syndrome – Promoter regions – Small interfering RNA – Transcription factors

Introduction

Androgens are traditionally considered as male hormones with well-established roles in male physiology and prostate cancer. However, in the last decade, several genetic models and in vitro studies have proven that androgens acting through androgen receptors (AR) are critical for ovarian function and female fertility [16]. While excess androgen level leads to polycystic ovary syndrome (PCOS) [79], a certain amount of direct androgen actions through the androgen receptor (AR) are essential for normal ovarian function [10]. Thus, it is now believed that with respect to androgen actions in the ovary, balance is key [4]. To date, in addition to the global androgen receptor knockout (ARKO) mouse models [1113], AR has been knocked out specifically in different cell types along the hypothalamus-pituitary-gonadal (HPG) axis, namely granulosa cells (GCARKO) [14,15], theca cells (TCARKO) [16], oocyte (OoARKO) [15], pituitary (PitARKO) [17] and neurons (NeuroARKO) regulating the HPG axis [18]. All of these ARKO mouse models establish that the granulosa cells (GCs) of the ovary are the primary site of androgen actions in regulating normal follicular development and female fertility; while in hyperandrogenic conditions, neuroendocrine ARs play a major role in the development of PCOS [18]. Moreover, ex vivo [5,19], in vitro [2023] and clinical studies [10,2431] show that androgens are essential for follicle growth while simultaneously preventing follicular atresia. Despite these studies, how androgens regulate these follicular endpoints is poorly understood.

The major androgens in women, in descending order of serum concentration, are dehydroepiandrosterone sulphate (DHEAS), dehydroepiandrosterone (DHEA), androstenedione (A4), testosterone (T), and dihydrotestosterone (DHT) [32]. However, direct androgenic actions through the AR can only be mediated by T and DHT, as these are the only two androgens that bind to AR, the latter being more potent [33]. The rest of the androgens are more like androgen precursors that require conversion to T and/or DHT to exert androgenic effects. In the ovarian follicle and stroma, androgen synthesis primarily involves the conversion of cholesterol into pregnenolone (by CYP11A1) that is metabolized to DHEA (by CYP17A1) and then A4 (by 3β-HSD), the immediate precursor of testosterone [34]. A4 is converted into testosterone (17β-HSD), that can then be aromatized into estradiol (E2) by aromatase (CYP19a1) or reduced to DHT by 5α-reductase [34,35]. In women, 5α-reductase (SRD5A1 and A2) mRNA expression has been found in both PCOS and non-PCOS women and interestingly, this expression is higher in granulosa cells than in theca cells [36,37]. In studies determining androgen actions in the ovary, DHT, which is non-aromatizable, is primarily used [5,19,38,39]. This is because T can be aromatized to estradiol, making it difficult to differentiate whether the downstream effects are due to estrogenic or androgenic actions, thereby confounding the results.

Androgen actions are mediated by “nuclear” transcriptional signals or “extra-nuclear” kinase actions [4042]. Primary AR target genes are those at which AR occupies an androgen response element (ARE) on the promoter of a gene and regulates gene transcription. However, to date, very few ovarian genes have been identified as AR-ARE target genes and, intriguingly, there are no studies on the global impact of androgens on GC gene expression under normal conditions. Here we describe androgen-induced gene expression profiles in mouse GCs and provide molecular insight into the underlying mechanism of how androgens regulate the expression of these genes.

Importantly, this study also shows that androgens can regulate gene expression in an AR-ARE independent fashion, involving membrane-initiated androgen signaling [5,4345]. Previously [38], we have reported that androgens influence gene expression through post-translational histone modifications. We have shown [38] that H3K27me3 (tri-methyl lysine 27 histone3) which is a gene silencing mark [46] is a downstream target of androgen actions. Here using ChIP-seq studies with H3K27me3 antibody we identify the ovarian (GC-specific) genes and their enhancer regions that are regulated by androgen-induced modulation of H3K27me3 mark. H3K27me3 is regulated by Enhancer of Zeste Homologue 2 (EZH2), a histone methyltransferase that promotes tri-methylation of lysine 27. Androgens, through both the extra-nuclear and nuclear pathways, inhibit the activity of EZH2 as well as Ezh2 expression, respectively [38]. Epigenetic modulation of gene expression is dependent partly on the dynamic balance of histone methylation/demethylation on the enhancers and promoters. This is mediated by regulating the expression and activity of methylating and/or demethylating enzymes [47,48]. We demonstrate that in addition to inhibiting Ezh2 expression and EZH2 activity, androgens also induce the expression of a histone demethylase called Jumonji domain containing protein 3 (Jmjd3/Kdm6b), that is responsible for removing the H3K27me3 mark. We find that in GCs, androgen in a transcription-independent fashion, increases hypoxia-inducible factor 1 alpha (HIF1α) protein levels, which in turn induce Jmjd3 expression. This study not only provides a mechanistic understanding of the global impact of androgens in normal follicular development, but may also contribute to comprehending the effects of excess androgens as seen in disease conditions like PCOS.

Results

Effect of androgen on granulosa cell (GC) transcriptome

Global effects of androgens in GCs were elucidated by RNA-seq analysis in primary mouse GC cultures treated with media (control) or DHT. The overall similarity among samples was assessed by the Euclidean distance between samples (S1 Fig). DESeq2 analysis identified a total of 190 annotated significant differentially expressed ENSEMBL genes (DEGs). Out of these genes, 129 were upregulated and 61 were downregulated genes (FPKM>1). The global transcriptional change across the two groups compared (control vs DHT) is represented by a volcano plot in Fig 1A and hierarchical clustering of all the significant DEGs in control vs DHT treated GCs are shown in Fig 1B. The complete list of significant DEGs is presented in S1 Dataset. In silico analysis revealed that all of the DEGs have at least one or more ARE sequences in the promoter and/or distal (within 5Kb) region thereby suggesting that most of these genes are regulated directly by AR, rather than secondary effects of hormone exposure. Based on established ovarian functions, 4 DEGs- Bmp4, Angpt1, Mmp2 and Lhcgr were selected to further verify that AR, through direct ARE binding, regulates the expression of these genes. Fig 1C shows DHT treatment significantly increases mRNA abundance of Bmp4, Angpt1, Mmp2 and Lhcgr. Moreover, ChIP-qPCR studies with AR antibody show that DHT treatment increases AR binding to AREs located on the promoter region of the above-mentioned genes (Fig 1D).

Androgen-induced transcriptome analysis in primary mouse granulosa cells (GC).
Fig. 1. Androgen-induced transcriptome analysis in primary mouse granulosa cells (GC).
A: Volcano plot: Representing the global transcriptional change across the groups compared. Each data point in the scatter plot represents a gene. Genes with an adjusted P ≤ 0.05 and a log2 fold change ≥ 1 are indicated by red dots and represent up-regulated genes. Genes with an adjusted P ≤ 0.05 and a log2 fold change ≤ -1 are indicated by blue dots and represent downregulated genes. B: Heatmap of differentially expressed genes sorted by adjusted p-value by plotting their log2 transformed expression values in samples. C: Relative expression of Bmp4 (Bone morphogenetic factor 4), Angpt1 (Angiopoietin 1), Mmp2 (Matrix Metallopeptidase 2) and Lhcgr (Luteinizing hormone/Choriogonadotropin receptor) mRNA levels by quantitative PCR in primary mouse GCs treated with media or DHT (25nM for 24h). Data are displayed as means ± SEM (n = 3 experiments, for each experiment GCs isolated from 5 mice were pooled together) and normalized to Rpl19 (* P ≤ 0.05, vs media). D: Anti-AR ChIP-assay in mouse primary GC cultures treated with media (control) or DHT (25nM for 24h) showing AR binding to different ARE sequences (P1/P2/P3) on Bmp4 (-693/-383Kb from TSS), Angpt1 (-1074Kb from TSS), Mmp2 (-937/-753/-332Kb from TSS) and Lhcgr (-47Kb from TSS) promoter region. IgG represents non-specific antibody. Values represent percentage input. Data are displayed as mean ± SEM (n = 3 experiments, for each experiment GCs isolated from 10 mice were pooled together) and * P ≤ 0.05, vs media. E: Gene ontology analysis: Gene ontology terms of significantly enriched pathways with an adjusted P-value ≤ 0.05 in the differentially expressed gene sets.

Gene ontology (GO) enrichment analysis (Fig 1E) of the DEGs revealed angiogenesis/tie pathway, cholesterol and lipid metabolism, steroid biosynthesis, oxidation-reduction process, ovarian follicle development and positive regulation of cell proliferation as some of the primary biological processes to be significantly affected by DHT treatment. Specific genes that are upregulated or downregulated in these pathways following androgen treatment are further shown in Fig 1E. These pathways/genes were previously not known to be regulated by androgens and, thereby, this study reveals that androgens affect a wide range of biological processes critical for normal follicular development and thus, ovarian function.

Androgens significantly modulate H3K27me3 mark on gene promoters and enhancers

Previously [38] we have shown that androgens decrease the H3K27me3 mark in GCs. To evaluate the impact of androgens on genome-wide distribution of H3K27me3 landscape, we performed ChIP with the H3K27me3 antibody followed by high-throughput sequencing in control vs. DHT treated GCs.

Total H3K27me3 peaks modulated by DHT

The analysis of sequencing reads revealed 16,345 H3K27me3 peaks in control and 3975 H3K27me3 peaks in DHT treated samples: a 75% reduction in peaks in the DHT treated samples. Fig 2A shows a heat map of genome wide H3K27me3 peaks in GCs from control and treatment groups. Each row in the heatmap corresponds to a H3K27me3 signal peak identified from either controls or DHT treated samples. The normalized ChIP-seq read densities around the peaks are shown with the summits of the peaks in the middle, along with +/-10kb flanking regions. Of 16,345 H3K27me3 peaks in control samples, 5513 peaks were within gene bodies while 153 peaks were in promoter regions. In contrast, there were only 1389 H3K37me3 peaks in the gene body and 54 peaks in the promoter region in the DHT treated samples. S2 Fig represents the number of H3K27me3 peaks overlapping different genomic annotations in the control and treatment (DHT) group.

Genes associated with androgen-induced decrease in H3K27me3 mark in the gene body and overlapping with promoters and distal enhancers involving long-range regulation of gene expression.
Fig. 2. Genes associated with androgen-induced decrease in H3K27me3 mark in the gene body and overlapping with promoters and distal enhancers involving long-range regulation of gene expression.
A: Heat maps showing the read density change along the peak regions for the 16345 control peaks and the 3975 treatment peaks. B: H3K27me3 signals in control and DHT treatment groups along the gene body of Fshr- follicle stimulating hormone receptor, Cyp19a1- aromatase, Lhcgr- luteinizing hormone/Choriogonadotropin receptor, Runx1- runt-related transcription factor 1, Egfr–epidermal growth factor receptor and Smad3- Mothers against decapentaplegic homolog 3. Log2 fold change was calculated as log2{(H3K27me3 control signal)/ (H3K27me3 treatment signal)}. Regions along the gene body with higher signals in the control group are represented as positive value- blue peaks and regions where signal was higher in the treatment group are shown as negative value-red peaks. C: Heatmap showing H3K27me3 peak counts in control and DHT-treated samples for 28 differentially expressed genes identified by comparing genes containing H3K27me3 peaks in the gene body with DEGs from the RNA-seq data. D: Average number of H3K27me3 peaks per 50bp overlapping with the promoter regions of 160 genes in the control samples and 55 genes in the DHT-treated samples. E: Degree analysis of enhancer-gene interactions for genes associated with significant decrease in H3K27me3 peaks in their enhancer regions with respect to DHT treatment. F: Distance between the promoters of genes and their corresponding enhancers that have decreased H3K27me3 signal by DHT treatment. G: Enriched transcription factor binding motifs in distal enhancers of genes associated with decrease in H3K27me3 signal with DHT treatment. E-values were < 0.1 and indicate the probabilities of observing the enrichment from random control DNA sequences. For each transcription factor, the upper motif logo corresponds to the consensus motif based on HOCOMOCO database and the lower motif logo corresponds to the observed sequence motifs that are enriched in linked distal enhancers.

DHT-induced modulation of H3K27me3 peaks in gene bodies

To further analyze the influence of androgen-modulated epigenetic dynamics on genes, we examined genes with gene bodies overlapping with H3K27me3 peaks. We identified 3144 genes in control and 1146 genes in DHT treated GCs with H3K27me3 peak signal across the gene body. Comparison of these two gene sets revealed that 2462 genes exclusively had H3K27me3 peak signal across the gene body in the control but not in the DHT-treated samples (S2 Dataset). Fig 2B demonstrates log2-fold change of H3K27me3 peaks overlapping with gene bodies of six representative genes (Fshr, Cyp19a1, Lhcgr, Runx1, Egfr and Smad3) that are known to play critical roles in ovarian function. For each gene, the H3K27me3 mark in control and treatment signals along the gene body was calculated by dividing each gene into 1000bp windows. The number of reads falling under each 1000bp window were considered the H3K27me3 signal in that window and log2 fold change of H3K27me3 signals along the gene body was calculated. Results show that DHT-treatment significantly lowers the H3K27me3 signal in all of the genes.

Subsequently, we compared the list of genes containing H3K27me3 peaks in their gene bodies with the list of DEGs from the RNA-seq data. We found 28 genes (22-upregulated and 6-downregulated genes) that were both differentially expressed and overlapped with H3K27me3 peaks, suggesting that the condition-specific epigenetic landscape of H3K27me3 may be related to the transcriptional variation of these genes. Fig 2C is a heat map representing the number of H3K27me3 peaks in the gene body (TSS to TES) for the 28 DEGs. Most of the upregulated genes had significantly lower levels of H3K27me3 marks while the downregulated genes had higher H3K27me3 marks in DHT-treated GCs than in controls. This shows that in addition to AR-ARE, the expression of these genes may also be regulated by androgen-induced H3K27me3 modulation.

DHT-induced modulation of H3K27me3 peaks in the promoter region

Further analysis revealed that there were 160 genes in control and only 55 genes in the DHT treated samples with H3K27me3 peaks located specifically in the gene promoter regions. Fig 2D (Left panel) represents the average number of H3K27me3 peaks per 50bp in the promoter region (TSS+/-1KB) for the 160 genes in the control group and corresponding average peaks for the same genes in the treatment group. Similarly, Fig 2D (right panel) represents the average number of H3K27me3 peaks per 50bp in the promoter region (TSS+/-1KB) for the 55 genes in the treatment group and corresponding average peaks for the same genes in the control group. The list of all the genes with promoters overlapping with H3K27me3 peaks in the control and treatment groups is provided in the supplemental data (S3 Dataset). Considering the complex regulatory activities in promoter regions, these androgen-induced differential H3K27me3 peaks located in the promoters may play pivotal roles in regulating the transcriptional levels of the corresponding genes.

DHT-induced modulation of H3K27me3 peaks in the enhancer region

Since large portions of the mouse genome are non-coding regions with numerous enhancers widely spread [49], we further extended our analysis to non-coding regions and focused on distal enhancers that have long-range chromatin interactions with promoters and may play a crucial role in controlling the expression of genes. While enhancers act through the binding of transcription factors just like promoters, their location greatly vary from the transcription start site (TSS) of the gene they regulate. Moreover, while a single enhancer can influence the expression of multiple genes, a single gene can be regulated by multiple enhancers. Thus, we determined enhancer-gene pairs for the 2462 genes in which the H3K27me3 signal peaks were significantly decreased by DHT treatment (genes with exclusive H3K27me3 peaks in the control group). For these genes, the chromatin interaction data including Hi-C and Capture-C were used to find potential enhancers (Bioinformatics analysis for ChIP-seq, S1 Text). For each gene, the H3K27me3 in the gene body and H3K27me3 level in each of its potential enhancers were calculated. We found 1380 genes where DHT treatment lowered the H3K27me3 signal (S5 Dataset). For enhancer–gene pair, the correlation between the gene body H3K27me3 level and enhancer H3K27me3 level was calculated and only positively correlated enhancer–gene pair (Pearson correlation > 0.4) were selected. Results show 3447 enhancer–gene pairs. Next, we determined the number of enhancers that regulate the same gene (Fig 2E). Results show that 45% of these genes are regulated by only 1 enhancer region while 21% of the genes are regulated by 2 enhancers. Furthermore, we calculated the distance between the promoters and their corresponding enhancers that have decreased H3K27me3 signal by DHT treatment. Fig 2F shows the distance analysis of the enhancer–gene interactions for the genes that show DHT-induced decrease in H3K27me3 levels. 33% of the enhancer–gene pairs that show decreased H3K27me3 with DHT treatment have 0 to 100KB distance between the gene and the enhancer. These analyses show that androgens can modulate gene expression not only by reducing the H3K27me3 mark in the promoter region of genes but also in distal enhancers.

Moreover, comparing the chromatin contact maps (ChIP-seq data) with the androgen-induced DEGS revealed 186 enhancers whose H3K27me3 levels were significantly negatively correlated (Pearson correlation < -0.4) with the expression of 99 DEGs, out of which 66 were upregulated and 33 were downregulated genes (S6 Dataset) across samples of controls and DHT treated GCs. This highlights the importance of complex long-range multi-enhancer regulation of AR regulated genes in the ovarian GCs.

Motif analysis of the enhancers

Given that H3K27me3 is a gene repressive mark, it is likely that the androgen-induced decrease of H3K27me3 allows specific transcription factors to bind to these enhancer regions. We analyzed the enhancer regions linked with H3K27me3 peaks for motif enrichment using MEME-ChIP as described in bioinformatics analysis for ChIP-seq, (S1 Text). Four transcription factors (TFs), FOXJ3 (forkhead box j3; p-value 1.44e-04 and q-value 1.49e-01), MAZ (MYC associated zinc finger protein; p-value 6.85e-08 and q-value 7.05e-05), SALL1 (spalt like transcription factor 1; p-value 2.20e-04 and q-value 9.90e-02) and SMAD3 (p-value 3.20e-05 and q-value 3.32e-02) with E-value < 0.1 were identified as candidate factors associated with epigenetic changes in distal enhancers (Fig 2G).

Androgens through the PI3K/Akt pathway, in a transcription-independent fashion, increase HIF1α protein levels, which in turn induce the expression of JMJD3 / KDM6B in mouse granulosa cells

Next, we determined the underlying mechanism by which androgens modulate H3K27me3. We have reported [38] that androgens inhibit the expression and activity of the histone methyltransferase EZH2, that promotes the H3K27me3 epigenetic mark. Here we wanted to determine if androgens have any effect on the expression of histone demethylases. Results (Fig 3A) show that 24h of DHT (25nM) treatment specifically induces the expression of a Jumonji domain containing demethylase called Jmjd3 (Kdm6b). There was no effect of DHT on the expression of other Jumonji domain containing demethylases namely, Jmjd1a (Kdm3a), Jmjd2b (Kdm4b), Jarid1b (Kdm5b), Jmjd2a (Kdm4b) and Jarid2. Intriguingly, Jmjd3 specifically demethylases H3K27me3 and therefore we further explored the mechanism by which androgens regulate the expression of Jmjd3.

Androgens through HIF1α regulate JMJD3 expression.
Fig. 3. Androgens through HIF1α regulate JMJD3 expression.
A: Relative expression of Jumonji histone demethylase mRNA levels by quantitative PCR in mouse primary GC cultures treated with media containing vehicle (control) or DHT (25nM for 24h). Data are displayed as mean ± SEM (n = 3 experiments, for each experiment GCs isolated from 5 mice were pooled together) and normalized to Rpl19 (* P ≤ 0.05, vs media). B: siRNA-mediated knockdown of Hif1α in mouse primary GC cultures. C: Effect of siRNA-mediated knockdown of Hif1α on Jmjd3 mRNA levels in mouse primary GC cultures treated with media containing vehicle (control) or DHT. Data are displayed as mean ± SEM (n = 3 experiments, for each experiment GCs isolated from 5 mice were pooled together) and normalized to Rpl19 (* P ≤ 0.05, vs media). D: Anti-HIF1α ChIP-assay in mouse primary GC cultures treated with media containing vehicle (control) or DHT (25nM for 24h) showing HIF1α binding to two (P1/P2) HRE sequences (-0.2/+4.2Kb from TSS) on Jmjd3 promoter region. IgG represents non-specific antibody and “Nsp” represents non-specific primers. Values represent percentage input. Data are displayed as mean ± SEM (n = 3 experiments, for each experiment GCs isolated from 10 mice were pooled together) and * P ≤ 0.05, vs media. E: HIF1α protein levels in mouse primary GC cultures treated with media containing vehicle (control) or DHT (25nM for 24h) in presence of flutamide (AR inhibitor, 100nM), LY294002 (PI3K inhibitor, 10μM) or cycloheximide (CHX, a protein translational inhibitor, 1μM). Epidermal Growth Factor, EGF (20ng/ml) was used as a positive control. F and G: Relative expression of Jmjd3 mRNA and protein levels (F) and H3K27me3 levels (G) in mouse primary GC cultures treated with media containing vehicle (control) or DHT (25nM for 24h) in presence of flutamide (AR inhibitor, 100nM) and LY294002 (PI3K inhibitor, 10μM). Data are displayed as mean ± SEM (n = 3 experiments, for each experiment GCs isolated from 5 mice were pooled together) and normalized to Rpl19 (P ≤ 0.05, * vs media and ** vs DHT). H: Proposed model of androgen-induced translation-dependent increase in HIF1α protein levels and its role in Jmjd3 expression. I: Effect of siRNA-mediated knockdown of Hif1α and Jmjd3 on H3K27me3 levels in mouse primary GC cultures treated with media containing vehicle (control) or DHT (25nM for 24h).

In silico analysis of the Jmjd3 promoter revealed the presence of HRE (Hif1α response element) regions (S3A Fig). Interestingly, studies in mouse and humans show that HIF1α can induce Jmjd3 both under normal conditions and hypoxic stress [50]. Therefore, to determine if HIF1α plays any role in DHT-induced expression of Jmjd3 in mouse GCs, we knocked down the expression of Hif1α with Hif1α -specific siRNA (Fig 3B) in primary mouse GC culture. siRNA-mediated knockdown of Hif1α completely blocked the DHT-induced increase of Jmjd3 mRNA (Fig 3C) and JMJD3 protein levels (Fig 3B) in mouse GCs. Furthermore, ChIP studies with HIF1α antibodies (Fig 3D) show increased binding of HIF1α on one of the two HRE sequences on the Jmjd3 promoter region with DHT treatment. This establishes that androgens, through HIF1α, mediate the expression of Jmjd3 in mouse GCs.

We further wanted to understand how androgens regulate HIF1α. In prostate cancer, it has been reported that androgens acting through the PI3K/Akt pathway, in a transcription-independent fashion, increase HIF1α protein levels [51]. Consequently, we and others have also reported that DHT treatment activates the PI3K/Akt pathway in GCs [6,19,38]. Results show that similar to prostate cancer, in mouse GCs, DHT stimulation does not affect Hif1α mRNA levels (Figs 3B and S3B) but increases HIF1α protein levels (Fig 3B and 3E). This increase in HIF1α protein by DHT stimulation is blocked by flutamide (AR inhibitor), LY294002 (PI3K inhibitor) and cycloheximide (CHX, a protein translational inhibitor) (Fig 3E). Moreover, CHX-chase studies with DHT revealed no difference in HIF1α protein degradation (S3C Fig), indicating that DHT increases HIF1α protein levels through a translation-dependent mechanism. Notably, inhibition of AR and the PI3K pathways attenuated DHT-induced increase in Jmjd3 mRNA (Fig 3F), JMJD3 protein levels (Fig 3F) and JMJD3 enzymatic activity (S3D Fig) as well as the global decrease in the H3K27me3 mark (Fig 3G) in mouse GCs. Thus, these studies show that androgens, through PI3K/Akt signaling, in a translation-dependent pathway enhance HIF1α protein levels that in turn bind to HRE sequences on the Jmjd3 promoter region and induce the expression of Jmjd3 in mouse GCs (Fig 3H).

Furthermore, to directly establish the AR-HIF1α-JMJD3 pathway to the downstream modulation of H3K27me3 mark, we knocked down Jmjd3 and Hif1α by siRNAs and determined H3K27me3 levels (Fig 3I), as well as performed ChIP assays with H3K27me3 antibody on Lhcgr, Cyp19a1, Fshr and Runx1 promoter regions (S4A Fig). Results show that ablation of Jmjd3 and Hif1α expressions block the androgen-induced decrease of total H3K27me3 levels (Fig 3I) and the H3K27me3 mark in the promoter region of the above-mentioned genes (S4A Fig).

Previous studies have shown that hypoxia alone or Clioquinol, a Hif1α activator under normoxia, can induce Jmjd3 expression [50]. Therefore, to determine whether exogenous activation of HIF1α in absence of AR can rescue Jmjd3 expression and H3K27me3 levels, we knocked down Ar by LNA-siRNA in primary mouse GC cultures and treated the cells with/without Clioquinol (50μM) in presence or absence of DHT (S5 Fig). Similar to previous studies, Clioquinol induces JMJD3 protein and lowers H3K27me3 level in mouse primary GCs (S5A Fig). However, in absence of AR, while Clioquinol completely rescues JMJD3 level, the downstream effect on H3K27me3 level (decrease of H3K27me3) was only partial (S5B Fig). This suggests that androgen-induced modulation of the H3K27me3 mark is mediated through a synergistic regulation of the expression and activity of the two opposing enzymes, EZH2 and JMJD3 that specifically target tri-methylation of H3K27. In addition to H3K27me3 levels, we also determined H3K27ac levels, a well-recognized marker for active enhancers. H3K27ac levels were significantly elevated in GCs treated with DHT compared to controls (S4B Fig).

Androgen-induced modulation of H3K27me3 regulates expression of genes critical for ovarian function

Finally, as a proof of concept, we knocked down AR expression in vivo by injecting LNA-containing oligonucleotides targeting AR or non-specific control into the ovarian bursa of mice (Fig 4A). Loss of AR expression in vivo significantly increases the H3K27me3 mark in the GCs that corresponds to increased expression of EZH2 and decreased levels of HIF1α and JMJD3 than in non-specific controls (Fig 4A). ChIP-PCR studies (Fig 4B) with H3K27me3 antibody also reveal higher level of H3K27me3 within the gene body of key ovarian genes, Lhcgr, Fshr, Cyp19a1 and Runx1. The downstream effect of increased H3K27me3 levels was further reflected by decrease in the mRNA levels of Lhcgr, Fshr, Cyp19a1 and Runx1 in the GCs treated with LNA-AR siRNA compared to non-specific control (Fig 4C).

<i>In vivo</i> knockdown of androgen receptors increases H3K27me3 mark and inhibits the expression of key ovarian genes.
Fig. 4. In vivo knockdown of androgen receptors increases H3K27me3 mark and inhibits the expression of key ovarian genes.
A: Protein levels of androgen receptor (AR), HIF1α, JMJD3, EZH2, H3K27me3, and H3 in GCs isolated from ovaries of mice injected in the ovarian bursa with LNA-locked Ar siRNA and non-specific (Nsp) siRNA control. B and C: Anti-H3K27me3 ChIP-assay showing H3K37me3 levels within the gene body of Lhcgr (+641/+740 bp from TSS), Fshr (+379/+488 bp from TSS), Cyp19a1 (+553/+653 bp from TSS) and Runx1 (+579/+693 bp from TSS) (B) and relative expression of mRNA of these genes (C) in GCs isolated from ovaries of mice (n = 15 mice) injected in the ovarian bursa with LNA-locked Ar siRNA and non-specific (Nsp) siRNA. IgG represents non-specific antibody. ChIP assay values represent percentage input and data are displayed as mean ± SEM (n = 3 ChIP assays, for each experiment GCs isolated from 5 mice were pooled together) and *P ≤ 0.05, vs non-specific siRNA. Relative expression of mRNA is displayed as mean ± SEM (n = 3 experiments, for each experiment GCs isolated from 5 mice were pooled together) and normalized to Rpl19 and *P ≤ 0.05, vs non-specific siRNA. D and E: H3K27me3, EZH2, JMJD3 and H3 protein levels (D) and relative expression of Fshr, Lhcgr and Cyp19a1 mRNA levels (E) in GCs isolated from ovaries of 24-25-week-old GC- specific ARKO (GCARKO) and wild type (WT) mice. Data are displayed as mean ± SEM (n = 4 mice) and normalized to Gapdh (*P ≤ 0.05 vs WT). F: Proposed model of androgen-induced modulation of H3K27me3 through regulation of Ezh2 and Jmjd3 expression and activity.

To further prove that the androgen/AR-induced modification of H3K27me3 is physiologically important, we determined H3K27me3 levels and expression of Lhcgr, Fshr and Cyp19a1 in GCs isolated from ovaries of 24-25-week-old GC-ARKO mice. Notably, we have shown previously [15,38] that GC-specific knockout of AR causes premature ovarian failure characterized by higher rate of atresia and fewer ovulated oocytes. Based on our present data it can be speculated that one of the reasons for the premature ovarian failure in the GC-ARKO mice is due to the loss of androgen-induced decrease in H3K27me3 mark that in turn regulates the expression of many key ovarian genes. Supporting this hypothesis, results show that knockout of ARs, specifically in the GCs of the ovary, indeed increases H3K27me3 and EZH2 levels (Fig 4D) with decrease in JMJD3 protein levels. Moreover, expression of Lhcgr, Fshr and Cyp19a1 are significantly lower (Fig 4E) in the GCs isolated from the ovaries of GC-ARKO animals compared to wild type.

Discussion

In the ovary, androgens are not merely a substrate for estrogen synthesis, but direct androgen actions through the ARs are critical for normal follicular development and female fertility [2,4]. However, there is a dearth of knowledge about the genes and biological pathways regulated by androgens, which is a significant limitation towards understanding how androgens regulate follicular growth and function. This study for the first time provides three critical insights about androgen actions in the ovary.

First, we have identified a large number of genes and biological processes that were not formerly known to be regulated directly by androgens in GCs. Results show that genes like Bmp4 [52], Lhcgr [53], Adamts4 [54], Ptgds4 and Mmp2 [5557], that are critical for follicular function are AR-induced genes. Previously, we [5] and others [6,19] have reported that androgens primarily maintain normal follicular development by regulating pre-antral to antral follicle transition by increasing FSH receptor levels and prevent follicular atresia. However, our gene expression data now clearly show that androgens have a much far-reaching impact on follicular function. For example, the angiopoietin-tie pathway, many of the genes of which we find to be directly induced by androgens, are not only vital for the remodeling and maturation of the developing vasculature that helps in the formation of dominant follicle/s but also play an essential role in non-angiogenic functions like follicular survival and steroidogenesis [58,59]. Inhibition of ANGPT1 increases follicular atresia [60] and the follicles which undergo early atresia show a higher ANGPT2:ANGPT1 ratio [6163]. This is consistent with our results—DHT treatment increases Angpt1 expression and lowers Angpt2 transcript levels. ANGPT2 is considered a natural antagonist of ANGPT1 [64,65]. The latter induces the phosphorylation of Tie2, which subsequently transduces a biological effect while ANGPT2 binds to Tie2 with the same affinity as ANGPT1 but does not phosphorylate the receptor [64,65]. Therefore, it is likely that the androgen-induced regulation of the angiopoietin–tie pathway is another mechanism by which androgens prevent follicular atresia. Notably, the fact that androgen treatment directly induces genes in the cholesterol-lipid metabolism, steroid biosynthesis and oxidation-reduction pathways, which are some of the basic biological processes involved in maintaining normal follicular development [6668], further highlights the importance of androgens in follicular development. Given that preovulatory androgens are essential for normal ovarian function and female fertility, while excess androgen is one of the well-established underlying causes for PCOS, we propose that there exists a critical balance between the essentiality of androgens for normal follicular function and the detrimental effects seen in hyperandrogenic conditions. For example, a number of genes like Lhcgr [69], Angpt1 [70] and Mmp2 [71], that were upregulated following androgen treatment (RNA-seq data), not only play critical role in follicular development but have also been shown to be elevated in PCOS patients.

Moreover, androgen treatment also resulted in downregulation of 61 genes. Interestingly, comparison of the RNA-seq and ChIP-seq dataset revealed that out of all the downregulated genes, only 6 genes had higher H3K27me3 mark (Fig 2C). This suggests that androgen-induced downregulation of genes may be a secondary effect of androgen treatment. For example, we have reported previously [38] that androgens induce the expression of miR-101 that in turn downregulates the expression of Ezh2. Another example is androgen-induced expression of miR-125b that decreases the expression of pro-apoptotic proteins [5].

Second, we have performed ChIP-seq analysis in GCs to determine the DHT-induced changes in H3K27me3, which is a gene silencing mark. Control of gene expression is exerted at a number of levels, one of which is the accessibility of genes and their controlling elements to the transcription machinery. Accessibility is dictated broadly by the degree of chromatin compaction, which is influenced in part by post-translational histone modifications. Our results highlight an important concept: in GCs, in addition to the genomic actions of androgens through the “classical” AR-ARE binding, androgen-induced decrease in H3K27me3 mark is another avenue through which androgens can regulate gene expression. The fact that we identified only 190 DEGs (from RNA-seq study) that are directly regulated by androgens in contrast to 2462 genes (from ChIP-seq) that specifically had lower H3K27me3 mark in DHT-treated GCs, clearly shows that androgen-induced modulation of H3K27me3 mark has a much broader impact than the direct effects of androgens on GC function. On the basis of our present study, we propose that in GCs, androgens prime the promoter and/or enhancer regions of genes by lowering the H3K27me3 mark, that enables other transcription factors to induce the expression of these genes. In fact, we have reported previously [38] that Runx1, a gene critical for ovulation, is one such downstream target and androgens remove the H3K27me3-repressive mark from the Runx1 promoter. This enables the hCG-induced transcription machinery to access the Runx1 promoter region leading to increased expression of Runx1. Intriguingly, previous studies have reported that androgen treatment increases the expression of genes like Fshr [6,7275] and Cyp19a1 [1,76,77] that are critical for follicular development. However, there was no evidence that these genes are direct targets of AR-ARE mediated actions. Prior to this study, it was not known how androgens regulated the expression of these critical ovarian genes to promote ovarian function and female fertility, in general. We now show that androgen treatment significantly lowers the H3K27me3 mark in the gene body of Fshr and Cyp19a1 which provides a mechanistic explanation of how androgens, independent of AR-ARE interaction, through H3K27me3 modulation may influence the expression of these genes. Moreover, some of the genes with lower H3K27me3 mark on the gene body/enhancer regions following DHT treatment, like Cyp19a1 [78], Adamts15 [79] Casp7 [80], Erbb4 [81] and Lepr [82] have been reported to be elevated and/or associated with PCOS.

Third, our studies highlight that in GCs, androgens modulate the H3K27me3 mark by balancing the expression and activity of two opposing epigenetic enzymes, EZH2 and JMJD3. While on one hand androgens inhibit EZH2 [38], which causes the tri-methylation of H3K27, androgens also induce the expression of Jmjd3, which is a histone demethylase that specifically removes the tri-methylation mark from H3K27 (Fig 4F). Moreover, in addition to the ‘genomic’ effects of AR, it is well-established that androgens can also induce transcription-independent ‘non-genomic’ effects [4042,45]. Our studies not only establish HIF1α as one of the downstream targets of non-genomic androgen actions, but also demonstrate that androgens indirectly induce the expression of Jmjd3 in GCs. Studies [83] in rat models show that Hif1α is expressed in the GCs throughout follicular development (from primary to pre-ovulatory follicles). Moreover, in pre-ovulatory follicles, Hif1α gene expression has been reported to be regulated by the actions of progesterone–progesterone receptor [84]. It is possible that dependent on the stage of follicular development, HIF1α may be regulated by different steroid hormones. For example, based on AR expression level at different follicular stages [1,85] as well as studies on androgenic effects during follicular development [4], it is believed that AR-mediated actions are predominantly in the pre-antral and small antral follicles and these effects diminish as the follicle develops to large antral/pre-ovulatory stage. Therefore, it can be speculated that in the pre-antral and small antral stages, androgens regulate HIF1α protein levels and following the LH surge, progesterone becomes the primary regulator of Hif1α. Further studies are needed to establish this hypothesis. Given that androgens increase HIF1α protein levels, it is likely that in addition to Jmjd3 expression, other HIF1α-target genes also get induced following androgen treatment. Intriguingly, a large number of upregulated genes in our RNA-seq data are HIF1α target genes. For example, Bmp4 [86,87], Angpt1 [88], Mmp2 [89,90], Has2 [91], Pcsk9 [92], Erg (ETS transcription factor) [93,94] and Ifitm1 [95] are some of the genes that have previously been reported to be HIF1α target genes. Studies [96] in cancer cells have reported significant crosstalk between AR and HIF1α. Co-immunoprecipitation assays have confirmed a direct interaction between AR and HIF1α, and ChIP analysis showed HIF1α interacts with the AR in genes involved in prostate cancer [96]. Similar interactions may also occur in the GCs.

Interestingly our studies also show that in addition to lowering the gene repressive H3K27me3 mark, androgens increase the gene activating H3K27ac mark. This is in accordance with previous studies that show reciprocal changes of H3K27ac and H3K27me3 modifications at the promoter regions of genes [97,98]. Moreover, EZH2 (inhibited by androgens) may function as an “on-off” switch, that acts not only as a critical epigenetic repressor, but also as an activator for specific downstream targets [99]. The EZH2 molecular switch functions mainly between H3K27me3 repressive mark and H3K27ac positive mark. EZH2 antagonizes CBP/p300 actions (primary enzyme for acetylation of lys-27 on H3) and inactivation or decrease of EZH2 increases CBP/p300-mediate acetylation of H3K27 [98,100]. Therefore, it is possible that in GCs, androgens by inhibiting Ezh2 and inducing Jmjd3 expression lower H3K27me3 mark and also simultaneously increase the H3K27ac mark. Further studies are needed not only to establish this hypothesis but also to determine the H3K27ac pattern with respect to gene expression in GCs following androgen treatment.

In summary, given the role of androgens in female fertility and women’s health in general, results of this study provide a global perception of androgen effects in follicular function and insights into the androgen-induced molecular mechanisms responsible for normal ovarian physiology as well as for disease conditions like PCOS.

Materials and methods

Details of all experimental methods are provided in the supplemental information (S1 Text).

Ethics statement

Mouse studies were performed in accordance with the guidelines for the care and use of laboratory animals and were approved by the Institutional Animal Care and Use Committee (IACUC) at MSU under the approval number PROTO202000156.

Animals and cell culture

Unless otherwise mentioned, mouse experiments were performed in 8–9 week old C57BL/6J mice (The Jackson Laboratories). Collection and culture of mouse GCs were performed as described previously [5,38,44,98,101103]. Different concentrations of DHT (5nM, 10nM and 25nM) were used to determine the effect of androgen on HIF1α, JMJD3 and H3K27me3 levels (S6A Fig) as well as gene expression of Bmp4, Angpt1, Lhcgr and Mmp2 (S6B Fig). All DHT concentrations showed similar results and based on our previous studies [38,45] 25nM DHT concentration was selected. DHT instead of testosterone was used in order to avoid misinterpretation of results due to aromatization of testosterone to estradiol.

siRNA knockdown experiments

siRNA-mediated knockdown experiments in primary mouse GCs [5,98,103] and ovarian bursal injections [5,38] were performed as previously described.

Chromatin immunoprecipitation (ChIP) assay

ChIP was performed as previously described [5,38,98,103,104] and List of ChIP primers (S4 Dataset) and antibodies (S7A Fig) are provided in the supplemental data.

RNA isolation, RNA-seq and Bioinformatics analysis

Total RNA isolation, library construction and RNA-sequencing services were carried out by Genewiz, Inc. Details of the RNA isolation, RNA-seq and bioinformatics analysis of RNA-seq data are provided in the supplemental material.

A list of all the differentially expressed genes is shown in S1 Dataset. The RNAseq data is available in the Gene Expression Omnibus GSE152727 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE152727).

Chromatin immunoprecipitation-sequencing (Chip-seq)

Chromatin isolation, chromatin shearing, ChIP, library preparation and Bioanalyzer QC was performed by EpiGentek, details of which are provided in the supplemental material. The ChIPseq data is available in the Gene Expression Omnibus GSE152791 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE152791).

Supporting information

S1 Fig [tif]
Sample similarity assessment: The overall similarity among samples were assessed by the euclidean distance between samples.

S2 Fig [tif]
Different genomic annotations associated with genome wide H3K27me3 peaks in granulosa cells treated with (treatment group) or without (control group) DHT (25nM for 24h).

S3 Fig [hre]

S4 Fig [nsp]

S5 Fig [tif]
Effect of Clioquinol, a HIF1α activator in presence/absence of AR on JMJD3 and H3K27me3 levels.

S6 Fig [tif]

S7 Fig [tif]

S1 Text [docx]
Materials and Methods.

S1 Dataset [xlsx]
List of differentially expressed genes (control vs DHT).

S2 Dataset [xlsx]
Genes with H3K27me3 peaks in the gene body of control and DHT-treated samples.

S3 Dataset [xlsx]
Genes with promoters overlapping with H3K27me3 peaks in control and DHT-treated samples.

S4 Dataset [xlsx]
ChIP primer list.

S5 Dataset [xlsx]
Genes with enhancers where H3K27me3 signals are lowered with DHT treatment.

S6 Dataset [xlsx]
Androgen-induced DEGS whose enhancers have significantly negative correlated H3K27me3 levels.


Zdroje

1. Walters KA. Role of androgens in normal and pathological ovarian function. Reproduction 2015; 149(4):R193–218. doi: 10.1530/REP-14-0517 25516989

2. Walters KA, Simanainen U, Gibson DA. Androgen action in female reproductive physiology. Curr Opin Endocrinol Diabetes Obes. 2016; 23(3):291–296. doi: 10.1097/MED.0000000000000246 26863277

3. Walters KA, Simanainen U, Handelsman DJ. Molecular insights into androgen actions in male and female reproductive function from androgen receptor knockout models. Hum Reprod Update. 2010; 16(5):543–558. doi: 10.1093/humupd/dmq003 20231167

4. Prizant H, Gleicher N, Sen A. Androgen actions in the ovary: balance is key. J Endocrinol. 2014; 222(3):R141–151. doi: 10.1530/JOE-14-0296 25037707

5. Sen A, Prizant H, Light A, Biswas A, Hayes E, Lee HJ, et al. Androgens regulate ovarian follicular development by increasing follicle stimulating hormone receptor and microRNA-125b expression. Proc Natl Acad Sci USA. 2014; 111(8):3008–3013. doi: 10.1073/pnas.1318978111 24516121

6. Franks S, Hardy K. Androgen Action in the Ovary. Front Endocrinol (Lausanne). 2018; 9:452. doi: 10.3389/fendo.2018.00452 30147675

7. Stener-Victorin E, Padmanabhan V, Walters KA, E Campbell RE, Benrick A, Giacobini P, et al. Animal models to understand the etiology and pathophysiology of polycystic ovary syndrome. Endocr Rev. 2020; 41(4):538–76. doi: 10.1210/endrev/bnaa010 32310267

8. Walters KA. Androgens in polycystic ovary syndrome: lessons from experimental models. Curr Opin Endocrinol Diabetes Obes. 2016; 23(3):257–263. doi: 10.1097/MED.0000000000000245 26866639

9. Walters KA, Bertoldo MJ, Handelsman DJ. Evidence from animal models on the pathogenesis of PCOS. Best Pract Res Clin Endocrinol Metab. 2018; 32(3):271–281. doi: 10.1016/j.beem.2018.03.008 29779581

10. Walters KA, Rodriguez Paris V, Aflatounian A, Handelsman DJ. Androgens and ovarian function: translation from basic discovery research to clinical impact. J Endocrinol. 2019; 242(2):R23–R50. doi: 10.1530/JOE-19-0096 31125975

11. Hu YC, Wang PH, Yeh S, Wang RS, Xie C, Xu Q, et al. Subfertility and defective folliculogenesis in female mice lacking androgen receptor. Proc Natl Acad Sci USA. 2004; 101(31):11209–11214. doi: 10.1073/pnas.0404372101 15277682

12. Shiina H, Matsumoto T, Sato T, Igarashi K, Miyamoto J, Takemasa S, et al. Premature ovarian failure in androgen receptor-deficient mice. Proc Natl Acad Sci USA. 2006; 103(1):224–229. doi: 10.1073/pnas.0506736102 16373508

13. Walters KA, Allan CM, Jimenez M, Lim PR, Davey RA, Zajac JD, et al. Female mice haploinsufficient for an inactivated androgen receptor (AR) exhibit age-dependent defects that resemble the AR null phenotype of dysfunctional late follicle development, ovulation, and fertility. Endocrinology. 2007; 148(8):3674–3684. doi: 10.1210/en.2007-0248 17463055

14. Walters KA, Middleton LJ, Joseph SR, Hazra R, Jimenez M, Simanainen U, et al. Targeted loss of androgen receptor signaling in murine granulosa cells of preantral and antral follicles causes female subfertility. Biol Reprod. 2012; 87(6):151. doi: 10.1095/biolreprod.112.102012 23115271

15. Sen A, Hammes SR. Granulosa cell-specific androgen receptors are critical regulators of ovarian development and function. Mol Endocrinol. 2010; 24(7):1393–1403. doi: 10.1210/me.2010-0006 20501640

16. Ma Y, Andrisse S, Chen Y, Childress S, Xue P, Wang Z, et al. Androgen Receptor in the Ovary Theca Cells Plays a Critical Role in Androgen-Induced Reproductive Dysfunction. Endocrinology. 2017; 158(1):98–108. doi: 10.1210/en.2016-1608 27841936

17. Wu S, Chen Y, Fajobi T, DiVall SA, Chang C, Yeh S, et al. Conditional knockout of the androgen receptor in gonadotropes reveals crucial roles for androgen in gonadotropin synthesis and surge in female mice. Mol Endocrinol. 2014; 28(10):1670–1681. doi: 10.1210/me.2014-1154 25157703

18. Caldwell ASL, Edwards MC, Desai R, Jimenez M, Gilchrist RB, Handelsman DJ, et al. Neuroendocrine androgen action is a key extraovarian mediator in the development of polycystic ovary syndrome. Proc Natl Acad Sci USA. 2017; 114(16):E3334–E3343. doi: 10.1073/pnas.1616467114 28320971

19. Laird M, Thomson K, Fenwick M, Mora J, Franks S, Hardy K. Androgen Stimulates Growth of Mouse Preantral Follicles In Vitro: Interaction With Follicle-Stimulating Hormone and With Growth Factors of the TGFbeta Superfamily. Endocrinology. 2017; 158(4):920–935. doi: 10.1210/en.2016-1538 28324051

20. Wang H, Andoh K, Hagiwara H, Xiaowei L, Kikuchi N, Abe Y et al. Effect of adrenal and ovarian androgens on type 4 follicles unresponsive to FSH in immature mice. Endocrinology. 2001; 142(11):4930–4936. doi: 10.1210/endo.142.11.8482 11606461

21. Hickey TE, Marrocco DL, Amato F, Ritter LJ, Norman RJ, Gilchrist RB, et al. Androgens augment the mitogenic effects of oocyte-secreted factors and growth differentiation factor 9 on porcine granulosa cells. Biol Reprod. 2005; 73(4):825–832. doi: 10.1095/biolreprod.104.039362 15972887

22. Hickey TE, Marrocco DL, Gilchrist RB, Norman RJ, Armstrong DT. Interactions between androgen and growth factors in granulosa cell subtypes of porcine antral follicles. Biol Reprod. 2004. 71(1):45–52. doi: 10.1095/biolreprod.103.026484 14973257

23. Murray AA, Gosden RG, Allison V, Spears N. Effect of androgens on the development of mouse follicles growing in vitro. J Reprod Fertil. 1998; 113(1):27–33. doi: 10.1530/jrf.0.1130027 9713373

24. Casson PR, Lindsay MS, Pisarska MD, Carson SA, Buster JE. Dehydroepiandrosterone supplementation augments ovarian stimulation in poor responders: a case series. Hum Reprod. 2000; 15(10):2129–2132. doi: 10.1093/humrep/15.10.2129 11006185

25. Barad D, Gleicher N. Effect of dehydroepiandrosterone on oocyte and embryo yields, embryo grade and cell number in IVF. Hum Reprod. 2006; 21(11):2845–2849. doi: 10.1093/humrep/del254 16997936

26. Barad DH, Gleicher N. Increased oocyte production after treatment with dehydroepiandrosterone. Fertil Steril. 2005; 84(3):756. doi: 10.1016/j.fertnstert.2005.02.049 16169414

27. Mamas L, Mamas E. Premature ovarian failure and dehydroepiandrosterone. Fertil Steril. 2009; 91(2):644–646. doi: 10.1016/j.fertnstert.2007.11.055 18321501

28. Hyman JH, Margalioth EJ, Rabinowitz R, Tsafrir A, Gal M, Alerhand S, et al. DHEA supplementation may improve IVF outcome in poor responders: a proposed mechanism. Eur J Obstet Gynecol Reprod Biol. 2013; 168(1):49–53. doi: 10.1016/j.ejogrb.2012.12.017 23312476

29. Sönmezer M, Ozmen B, Cil AP, Ozkavukçu S, Taşçi T, Olmuş H, et al. Dehydroepiandrosterone supplementation improves ovarian response and cycle outcome in poor responders. Reprod Biomed Online. 2009; 19(4):508–513. doi: 10.1016/j.rbmo.2009.06.006 19909591

30. Sunkara SK, Coomarasamy A. Androgen pretreatment in poor responders undergoing controlled ovarian stimulation and in vitro fertilization treatment. Fertil Steril. 2011; 95(8):e73–74. doi: 10.1016/j.fertnstert.2011.04.083 21601849

31. Gonzalez-Comadran M, Durán M, Solà I, Fábregues F, Carreras R, Checa MA, et al. Effects of transdermal testosterone in poor responders undergoing IVF: systematic review and meta-analysis. Reprod Biomed Online. 2012; 25(5):450–459. doi: 10.1016/j.rbmo.2012.07.011 22999555

32. Davison SL, Davis SR. Androgens in women. J Steroid Biochem Mol Biol. 2003; 85(2–5):363–366. doi: 10.1016/s0960-0760(03)00204-8 12943723

33. Burger HG. Androgen production in women. Fertil Steril. 2002; 77 Suppl 4:S3–5. doi: 10.1016/s0015-0282(02)02985-0 12007895

34. Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. 2011; 32(1):81–151. doi: 10.1210/er.2010-0013 21051590

35. Ghayee HK, Auchus RJ. Basic concepts and recent developments in human steroid hormone biosynthesis. Rev Endocr Metab Disord. 2007; 8(4):289–300. doi: 10.1007/s11154-007-9052-2 17926129

36. Jakimiuk AJ, Weitsman SR, Magoffin DA. 5alpha-reductase activity in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 1999; 84(7):2414–2418. doi: 10.1210/jcem.84.7.5863 10404813

37. Kayampilly PP, Wanamaker BL, Stewart JA, Wagner CL, Menon KM. Stimulatory effect of insulin on 5alpha-reductase type 1 (SRD5A1) expression through an Akt-dependent pathway in ovarian granulosa cells. Endocrinology. 2010; 151(10):5030–5037. doi: 10.1210/en.2010-0444 20810561

38. Ma X, Hayes E, Biswas A, Seger C, Prizant H, Hammes SR, et al. Androgens Regulate Ovarian Gene Expression Through Modulation of Ezh2 Expression and Activity. 2017; 158(9):2944–2954. doi: 10.1210/en.2017-00145 28666321

39. Xue K, Liu JY, Murphy BD, Tsang BK. Orphan nuclear receptor NR4A1 is a negative regulator of DHT-induced rat preantral follicular growth. Mol Endocrinol. 2012. 26(12):2004–2015. doi: 10.1210/me.2012-1200 23028064

40. Hammes SR, Levin ER. Minireview: Recent advances in extranuclear steroid receptor actions. Endocrinology. 2011. 152(12):4489–4495. doi: 10.1210/en.2011-1470 22028449

41. Hammes SR, Levin ER. Extranuclear steroid receptors: nature and actions. Endocr Rev. 2007. 28(7):726–741. doi: 10.1210/er.2007-0022 17916740

42. Levin ER, Hammes SR. Nuclear receptors outside the nucleus: extranuclear signalling by steroid receptors. Nat Rev Mol Cell Biol. 2016; 17(12):783–797. doi: 10.1038/nrm.2016.122 27729652

43. Sen A, De Castro I, Defranco DB, Deng FM, Melamed J, Kapur P, et al. Paxillin mediates extranuclear and intranuclear signaling in prostate cancer proliferation. J Clin Invest. 2012; 122(7):2469–2481. doi: 10.1172/JCI62044 22684108

44. Sen A, O’Malley K, Wang Z, Raj GV, Defranco DB, Hammes SR, et al. Paxillin regulates androgen- and epidermal growth factor-induced MAPK signaling and cell proliferation in prostate cancer cells. J Biol Chem. 2010; 285(37):28787–28795. doi: 10.1074/jbc.M110.134064 20628053

45. Sen A, Prizant H, Hammes SR. Understanding extranuclear (nongenomic) androgen signaling: what a frog oocyte can tell us about human biology. Steroids. 2011; 76(9):822–828. doi: 10.1016/j.steroids.2011.02.016 21354434

46. Viré E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature. 2006; 439(7078):871–874. doi: 10.1038/nature04431 16357870

47. Blum R. Stepping inside the realm of epigenetic modifiers. Biomol Concepts. 2015; 6(2):119–136. doi: 10.1515/bmc-2015-0008 25915083

48. Hyun K, Jeon J, Park K, Kim J. Writing, erasing and reading histone lysine methylations. Exp Mol Med. 2017; 49(4):e324. doi: 10.1038/emm.2017.11 28450737

49. Yue F, Cheng Y, Breschi A, Vierstra J, Wu W, Ryba T, et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature. 2014; 515(7527):355–364. doi: 10.1038/nature13992 25409824

50. Lee HY, Choi K, Oh H, Park YK, Park H. HIF-1-dependent induction of Jumonji domain-containing protein (JMJD) 3 under hypoxic conditions. Mol Cells. 2014; 37(1):43–50. doi: 10.14348/molcells.2014.2250 24552709

51. Mabjeesh NJ, Willard MT, Frederickson CE, Zhong H, Simons JW. Androgens stimulate hypoxia-inducible factor 1 activation via autocrine loop of tyrosine kinase receptor/phosphatidylinositol 3’-kinase/protein kinase B in prostate cancer cells. Clin Cancer Res. 2003; 9(7):2416–2425. 12855613

52. Nilsson EE, Skinner MK. Bone morphogenetic protein-4 acts as an ovarian follicle survival factor and promotes primordial follicle development. Biol Reprod. 2003; 69(4):1265–1272. doi: 10.1095/biolreprod.103.018671 12801979

53. Narayan P. Genetic Models for the Study of Luteinizing Hormone Receptor Function. Front Endocrinol (Lausanne). 2015; 6:152.

54. Richards JS, Hernandez-Gonzalez I, Gonzalez-Robayna I, Teuling E, Lo Y, Boerboom D, et al. Regulated expression of ADAMTS family members in follicles and cumulus oocyte complexes: evidence for specific and redundant patterns during ovulation. Biol Reprod. 2005; 72(5):1241–1255. doi: 10.1095/biolreprod.104.038083 15659705

55. Light A, Hammes SR. LH-Induced Steroidogenesis in the Mouse Ovary, but Not Testis, Requires Matrix Metalloproteinase 2- and 9-Mediated Cleavage of Upregulated EGF Receptor Ligands. Biol Reprod. 2015; 93(3):65. doi: 10.1095/biolreprod.115.130971 26203177

56. Carbajal L, Biswas A, Niswander LM, Prizant H, Hammes SR. GPCR/EGFR cross talk is conserved in gonadal and adrenal steroidogenesis but is uniquely regulated by matrix metalloproteinases 2 and 9 in the ovary. Mol Endocrinol. 2011; 25(6):1055–1065. doi: 10.1210/me.2010-0410 21454403

57. Jamnongjit M, Gill A, Hammes SR. Epidermal growth factor receptor signaling is required for normal ovarian steroidogenesis and oocyte maturation. Proc Natl Acad Sci USA. 2005; 102(45):16257–16262. doi: 10.1073/pnas.0508521102 16260720

58. Abramovich D, Rodriguez Celin A, Hernandez F, Tesone M, Parborell F. Spatiotemporal analysis of the protein expression of angiogenic factors and their related receptors during folliculogenesis in rats with and without hormonal treatment. Reproduction. 2009; 137(2):309–320. doi: 10.1530/REP-08-0130 18996974

59. Robinson RS, Woad KJ, Hammond AJ, Laird M, Hunter MG, Mann GE. Angiogenesis and vascular function in the ovary. Reproduction. 2009; 138(6):869–881. doi: 10.1530/REP-09-0283 19786399

60. Parborell F, Abramovich D, Tesone M. Intrabursal administration of the antiangiopoietin 1 antibody produces a delay in rat follicular development associated with an increase in ovarian apoptosis mediated by changes in the expression of BCL2 related genes. Biol Reprod. 2008; 78(3):506–513. doi: 10.1095/biolreprod.107.063610 17989359

61. Hayashi KG, Acosta TJ, Tetsuka M, Berisha B, Matsui M, Schams D, et al. Involvement of angiopoietin-tie system in bovine follicular development and atresia: messenger RNA expression in theca interna and effect on steroid secretion. Biol Reprod. 2003; 69(6):2078–2084. doi: 10.1095/biolreprod.103.017152 12954734

62. Wulff C, Wilson H, Largue P, Duncan WC, Armstrong DG, Fraser HM. Angiogenesis in the human corpus luteum: localization and changes in angiopoietins, tie-2, and vascular endothelial growth factor messenger ribonucleic acid. J Clin Endocrinol Metab. 2000; 85(11):4302–4309. doi: 10.1210/jcem.85.11.6942 11095472

63. Xu F, Stouffer RL. Local delivery of angiopoietin-2 into the preovulatory follicle terminates the menstrual cycle in rhesus monkeys. Biol Reprod. 2005. 72(6):1352–1358. doi: 10.1095/biolreprod.104.037143 15703373

64. Hanahan D. Signaling vascular morphogenesis and maintenance. Science. 1997. 277(5322):48–50. doi: 10.1126/science.277.5322.48 9229772

65. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science. 1997. 277(5322):55–60. doi: 10.1126/science.277.5322.55 9204896

66. Chang XL, Liu L, Wang N, Chen ZJ, Zhang C. The function of high-density lipoprotein and low-density lipoprotein in the maintenance of mouse ovarian steroid balance. Biol Reprod. 2017; 97(6):862–872. doi: 10.1093/biolre/iox134 29092018

67. Huang Q, Liu Y, Yang Z, Xie Y, Mo Z. The Effects of Cholesterol Metabolism on Follicular Development and Ovarian Function. Curr Mol Med. 2019; 19(10):719–730. doi: 10.2174/1566524019666190916155004 31526349

68. Lu J, Wang Z, Cao J, Chen Y, Dong Y. A novel and compact review on the role of oxidative stress in female reproduction. Reprod Biol Endocrinol. 2018; 16(1):80. doi: 10.1186/s12958-018-0391-5 30126412

69. Kanamarlapudi V, Gordon UD, Lopez Bernal A. Luteinizing hormone/chorionic gonadotrophin receptor overexpressed in granulosa cells from polycystic ovary syndrome ovaries is functionally active. Reprod Biomed Online. 2016; 32(6):635–641. doi: 10.1016/j.rbmo.2016.03.003 27061682

70. Scotti L, Parborell F, Irusta G, De Zuñiga I, Bisioli C, Pettorossi H, et al. Platelet-derived growth factor BB and DD and angiopoietin1 are altered in follicular fluid from polycystic ovary syndrome patients. Mol Reprod Dev. 2014; 81(8):748–756. doi: 10.1002/mrd.22343 24889290

71. Lewandowski KC, Komorowski J, O’Callaghan CJ, Tan BK, Chen J, Prelevic GM, et al. Increased circulating levels of matrix metalloproteinase-2 and -9 in women with the polycystic ovary syndrome. J Clin Endocrinol Metab. 2006; 91(3):1173–1177. doi: 10.1210/jc.2005-0648 16338908

72. Vendola KA, Zhou J, Adesanya OO, Weil SJ, Bondy CA. Androgens stimulate early stages of follicular growth in the primate ovary. J Clin Invest. 1998; 101(12):2622–2629. doi: 10.1172/JCI2081 9637695

73. Weil S, Vendola K, Zhou J, Bondy CA. Androgen and follicle-stimulating hormone interactions in primate ovarian follicle development. J Clin Endocrinol Metab. 1999 84(8):2951–2956. doi: 10.1210/jcem.84.8.5929 10443703

74. Weil SJ, Vendola K, Zhou J, Adesanya OO, Wang J, Okafor J, et al. Androgen receptor gene expression in the primate ovary: cellular localization, regulation, and functional correlations. J Clin Endocrinol Metab. 1998; 83(7):2479–2485. doi: 10.1210/jcem.83.7.4917 9661631

75. Vendola K, Zhou J, Wang J, Famuyiwa OA, Bievre M, Bondy CA. Androgens promote oocyte insulin-like growth factor I expression and initiation of follicle development in the primate ovary. Biol Reprod. 1999; 61(2):353–357. doi: 10.1095/biolreprod61.2.353 10411511

76. Fitzpatrick SL, Richards JS. Regulation of cytochrome P450 aromatase messenger ribonucleic acid and activity by steroids and gonadotropins in rat granulosa cells. Endocrinology. 1991; 129(3):1452–1462. doi: 10.1210/endo-129-3-1452 1651851

77. Tetsuka M, Hillier SG. Differential regulation of aromatase and androgen receptor in granulosa cells. J Steroid Biochem Mol Biol. 1997; 61(3–6):233–239. 9365195

78. Owens LA, Kristensen SG, Lerner A, Christopoulos G, Lavery S, Hanyaloglu AC, et al. Gene Expression in Granulosa Cells From Small Antral Follicles From Women With or Without Polycystic Ovaries. J Clin Endocrinol Metab. 2019; 104(12):6182–6192. doi: 10.1210/jc.2019-00780 31276164

79. Ambekar AS, Kelkar DS, Pinto SM, Sharma R, Hinduja I, Zaveri K, et al. Proteomics of follicular fluid from women with polycystic ovary syndrome suggests molecular defects in follicular development. J Clin Endocrinol Metab. 2015; 100(2):744–753. doi: 10.1210/jc.2014-2086 25393639

80. Salehi E, Aflatoonian R, Moeini A, Yamini N, Asadi E, Khosravizadeh Z, et al. Apoptotic biomarkers in cumulus cells in relation to embryo quality in polycystic ovary syndrome. Arch Gynecol Obstet. 2017; 296(6):1219–1227. doi: 10.1007/s00404-017-4523-5 28988321

81. Peng Y, Zhang W, Yang P, Tian Y, Su S, Zhang C, et al. ERBB4 Confers Risk for Polycystic Ovary Syndrome in Han Chinese. Sci Rep. 2017 7:42000. doi: 10.1038/srep42000 28195137

82. Li L, Lee KJ, Choi BC, Baek KH. Relationship between leptin receptor and polycystic ovary syndrome. Gene. 2013; 527(1):71–74. doi: 10.1016/j.gene.2013.05.074 23769971

83. Zhang ZH, Chen LY, Wang F, Wu YQ, Su JQ, Huang XH, et al. Expression of hypoxia-inducible factor-1alpha during ovarian follicular growth and development in Sprague-Dawley rats. Genet Mol Res. 2015; 14(2):5896–5909. doi: 10.4238/2015.June.1.7 26125789

84. Kim J, Bagchi IC, Bagchi MK. Signaling by hypoxia-inducible factors is critical for ovulation in mice. Endocrinology. 2009; 150(7):3392–3400. doi: 10.1210/en.2008-0948 19325003

85. Lenie S, Smitz J. Functional AR signaling is evident in an in vitro mouse follicle culture bioassay that encompasses most stages of folliculogenesis. Biol Reprod. 2009; 80(4):685–695. doi: 10.1095/biolreprod.107.067280 19074005

86. Wang J, Fu X, Yang K, Jiang Q, Chen Y, Jia J, et al. Hypoxia inducible factor-1-dependent up-regulation of BMP4 mediates hypoxia-induced increase of TRPC expression in PASMCs. Cardiovasc Res. 2015. 107(1):108–118. doi: 10.1093/cvr/cvv122 25824146

87. Wu DC, Paulson RF. Hypoxia regulates BMP4 expression in the murine spleen during the recovery from acute anemia. PLoS One. 2010; 5(6):e11303. doi: 10.1371/journal.pone.0011303 20585586

88. Dengler VL, Galbraith M, Espinosa JM. Transcriptional regulation by hypoxia inducible factors. Crit Rev Biochem Mol Biol. 2014; 49(1):1–15. doi: 10.3109/10409238.2013.838205 24099156

89. Wan R, Mo Y, Chien S, Li Y, Li Y, Tollerud DJ, et al. The role of hypoxia inducible factor-1alpha in the increased MMP-2 and MMP-9 production by human monocytes exposed to nickel nanoparticles. Nanotoxicology. 2011; 5(4):568–582. doi: 10.3109/17435390.2010.537791 21401309

90. Tsai SH, Huang PH, Hsu YJ, Peng YJ, Lee CH, Wang JC, et al. Inhibition of hypoxia inducible factor-1alpha attenuates abdominal aortic aneurysm progression through the down-regulation of matrix metalloproteinases. Sci Rep. 2016; 6:28612. doi: 10.1038/srep28612 27363580

91. Zhu G, Wang S, Chen J, Wang Z, Liang X, Wang X, et al. Long noncoding RNA HAS2-AS1 mediates hypoxia-induced invasiveness of oral squamous cell carcinoma. Mol Carcinog. 2017; 56(10):2210–2222. doi: 10.1002/mc.22674 28485478

92. Ding Z, Wang X, Liu S, Shahanawaz J, Theus S, Fan Y, et al. PCSK9 expression in the ischaemic heart and its relationship to infarct size, cardiac function, and development of autophagy. Cardiovasc Res. 2018; 114(13):1738–1751. doi: 10.1093/cvr/cvy128 29800228

93. Oikawa M, Abe M, Kurosawa H, Hida W, Shirato K, Sato Y. Hypoxia induces transcription factor ETS-1 via the activity of hypoxia-inducible factor-1. Biochem Biophys Res Commun. 2001; 289(1):39–43. doi: 10.1006/bbrc.2001.5927 11708773

94. Qiao N, Xu C, Zhu YX, Cao Y, Liu DC, Han X. Ets-1 as an early response gene against hypoxia-induced apoptosis in pancreatic beta-cells. Cell Death Dis. 2015; 6:e1650. doi: 10.1038/cddis.2015.8 25695603

95. Wan J, Ma J, Mei J, Shan G. The effects of HIF-1alpha on gene expression profiles of NCI-H446 human small cell lung cancer cells. 2009; J Exp Clin Cancer Res 28:150. doi: 10.1186/1756-9966-28-150 20003295

96. Horii K, Suzuki Y, Kondo Y, Akimoto M, Nishimura T, Yamabe Y, et al. Androgen-dependent gene expression of prostate-specific antigen is enhanced synergistically by hypoxia in human prostate cancer cells. Mol Cancer Res. 2007; 5(4):383–391. doi: 10.1158/1541-7786.MCR-06-0226 17426252

97. Katoh N, Kuroda K, Tomikawa J, Ogata-Kawata H, Ozaki R, Ochiai A, et al. Reciprocal changes of H3K27ac and H3K27me3 at the promoter regions of the critical genes for endometrial decidualization. Epigenomics. 2018; 10(9):1243–1257. doi: 10.2217/epi-2018-0006 30212243

98. Sinha N, Biswas A, Nave O, Seger C, Sen A. Gestational Diabetes Epigenetically Reprograms the Cart Promoter in Fetal Ovary, Causing Subfertility in Adult Life. Endocrinology. 2019; 160(7):1684–1700. doi: 10.1210/en.2019-00319 31150057

99. Pasini D, Malatesta M, Jung HR, Walfridsson J, Willer A, Olsson L, et al. Characterization of an antagonistic switch between histone H3 lysine 27 methylation and acetylation in the transcriptional regulation of Polycomb group target genes. Nucleic Acids Res. 2010; 38(15):4958–4969. doi: 10.1093/nar/gkq244 20385584

100. Xiong Y, Khanna S, Grzenda AL, Sarmento OF, Svingen PA, Lomberk GA, et al. Polycomb antagonizes p300/CREB-binding protein-associated factor to silence FOXP3 in a Kruppel-like factor-dependent manner. J Biol Chem. 2012; 287(41):34372–34385. doi: 10.1074/jbc.M111.325332 22896699

101. Ma X, Hayes E, Prizant H, Srivastava RK, Hammes SR, Sen A. Leptin-induced CART (Cocaine-and Amphetamine-Regulated Transcript) is a novel intra-ovarian mediator of obesity-related infertility in females. Endocrinology. 2016; en20151750.

102. Hayes E, Kushnir V, Ma X, Biswas A, Prizant H, Gleicher N, et al. Intra-cellular mechanism of Anti-Mullerian hormone (AMH) in regulation of follicular development. Mol Cell Endocrinol. 2016; 433:56–65. doi: 10.1016/j.mce.2016.05.019 27235859

103. Roy S, Gandra D, Seger C, Biswas A, Kushnir VA, Gleicher N, et al. Oocyte-Derived Factors (GDF9 and BMP15) and FSH Regulate AMH Expression Via Modulation of H3K27AC in Granulosa Cells. Endocrinology. 2018; 159(9):3433–3445. doi: 10.1210/en.2018-00609 30060157

104. Sinha N, Roy S, Huang B, Wang J, Padmanabhan V, Sen A. Developmental Programming: Prenatal Testosterone-induced Epigenetic Modulation and its Effect on Gene Expression in Sheep Ovary. Biol Reprod. 2020; 102(5):1045–1054. doi: 10.1093/biolre/ioaa007 31930385


Článek vyšel v časopise

PLOS Genetics


2021 Číslo 3
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#