Skip to main content
  • Research article
  • Open access
  • Published:

Identification of new progestogen-associated networks in mammalian ovulation using bioinformatics

Abstract

Background

Progesterone plays an essential role in mammalian ovulation. Although much is known about this process, the gene networks involved in ovulation have yet to be established. When analyze the mechanisms of ovulation, we often need to determine key genes or pathways to investigate the reproduction features. However, traditional experimental methods have a number of limitations.

Results

Data, in this study, were acquired from GSE41836 and GSE54584 which provided different samples. They were analyzed with the GEO2R and 546 differentially expressed genes were obtained from two data sets using bioinformatics (absolute log2 FC > 1, P < 0.05). This study identified four genes (PGR, RELN, PDE10A and PLA2G4A) by protein-protein interaction networks and pathway analysis, and their functional enrichments were associated with ovulation. Then, the top 25 statistical pathway enrichments related to hCG treatment were analyzed. Furthermore, gene network analysis identified certain interconnected genes and pathways involved in progestogenic mechanisms, including progesterone-mediated oocyte maturation, the MAPK signaling pathway, the GnRH signaling pathway and focal adhesion, etc. Moreover, we explored the four target gene pathways. q-PCR analysis following hCG and RU486 treatments confirmed the certain novel progestogenic-associated genes (GNAI1, PRKCA, CAV1, EGFR, RHOA, ZYX, VCL, GRB2 and RAP1A).

Conclusions

The results suggested four key genes, nine predicted genes and eight pathways to be involved in progestogenic networks. These networks provide important regulatory genes and signaling pathways which are involved in ovulation. This study provides a fundamental basis for subsequent functional studies to investigate the regulation of mammalian ovulation.

Background

Mammalian reproduction depends on the perfect ovulation of a fully differentiated oocyte capable of fertilisation. This process is triggered by hormonal release at the hypothalamic-pituitary-ovarian axis [1, 2]. Ovulation is initiated by gonadotropin luteinising hormone (LH). This is a complex process that includes follicle-wall degradation, vasodilation, vascular dynamics and inflammation [2]. In this context, it is important to elucidate the mechanism of mammalian ovulation required for effective reproduction.

Progesterone and its receptor (progesterone receptor, PGR) are both required in the ovulation process. The inhibition of progesterone synthesis by mifepristone is known to block ovulation in rats [2]. The anovulatory phenotype of the PGR-null mouse with a distinct defect in follicle-wall breakdown presents an opportunity to investigate molecular pathways. It is known that PGR controls the expression of a number of downstream target genes which regulate the ovulatory process [2,3,4,5]. Studies have identified certain progesterone-related factors including estradiol (E2), epidermal growth factor (Egf) [6], prostaglandin-endoperoxide synthase 2 (Ptgs2) [7], vascular endothelial growth factor (Vegfa) [8] and interleukin-6 (Il6) [9]. Furthermore, experiments using gene-null mice identified unruptured follicles and the failure of follicle-wall breakdown as the two causes of impaired ovulation. This analysis revealed novel gene signaling pathways which control the activity of PGR downstream to regulate ovulation, including PGR-Hif-Adamts1 (follicle-wall degradation), the PGR-Hif-Vegfa (vascular permeability) and PGR-Pparγ-Il6-Ptgs2 (inflammation) pathways [2]. However, the process in which these systematic signaling networks are involved the regulation of ovulation has not been identified.

Unlike traditional methods, bioinformatics analysis can explore multi-component and multi-target molecular networks. In study of ovarian primordial follicle assembly, much of the information derived from sequencing data needs further analysis, including a systems biology approach to identify gene expression networks [10,11,12,13,14,15]. This approach can indicate novel factors and potential targets that correlate with ovarian disease. Grøndah et al. [16] described the transcriptome associated with oocyte development by in silico microarray analysis, identified differentially expressed genes by bioinformatics analysis, and further validated the identified genes through conducting quantitative real-time polymerase chain reactions (q-PCR) analysis and confirming the functional categories. These studies suggested that bioinformatics analysis based on sequencing data provided not only a systematic understanding of complex biological processes, but also a promising and efficient method to explore physiological functions in follicle development [13]. Current research focusing on the molecular pathways and networks involved in ovulation remains insufficient, and further bioinformatics analysis is needed.

The present study aims to explore gene networks and predict putative regulatory factors involved in ovulation by using bioinformatics analysis. Our objectives are to: 1) identify the most common and specific differentially expressed genes through investigating the protein-protein interactions; 2) determine the key candidate genes by constructing biological gene networks involved in progesterone production during ovulation; 3) verify the candidate genes in progestogenic pathways. This study would improve the understanding of molecular functions by targeting specific pathways in the progestogenic networks, which might open new horizons into the investigation of the normal physiology of the ovaries.

Methods

Data acquisition and analysis

Data were acquired from the Gene Expression Omnibus (GEO) (http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/geo/) database. Two data sets, GSE54584 (this research clarified genetic alteration induced by the inhibition of cyclooxygenase or progesterone receptor) and GSE41836 (This study provided comprehensive information about regulated genes during late follicle development and ovulation induction), were contributed by Kenjiro et al. [17] and Cansu et al. [18], respectively. GSE41836 total contains 12 samples of cumulus oocyte complexes, among which only three samples with human Chorionic Gonadotropins (hCG) treatment were selected. GSE54584 total contains 18 samples, six samples including three samples with hCG treatment and three samples with RU486 (100 mg/kg) treatment in preovulatory were selected [17, 18]. These selected samples were analyzed by the GEO2R (http://0-www-ncbi-nlm-nih-gov.brum.beds.ac.uk/geo/geo2r/) with respect to the control groups (samples with same dose normal saline treatment). Multiple testing was corrected using the Benjamini-Hochberg procedure to obtain the adjusted P-value (P < 0.05). Fold changes in the expression of individual genes were calculated and the genes with absolute log2 FC > 1 were considered as differentially expressed genes (DEGs). Accordingly, DEGs determined from the selected data sets were identified as DEG41836 and DEG54584, the intersection of which was referred to as the overlapping DEGs.

Protein-protein interaction (PPI) network analysis

The Search Tool for the Retrieval of Interacting Genes (STRING, version 9.1) database (http://www.bork.embl-heidelberg.de/STRING/) is a pre-computed global resource biological database that includes comprehensive known and predicted protein interaction information. In this study, the STRING 9.1 online tool was used to screen the overlapping DEGs with a confidence score over 0.4. The overlapping DEGs with hCG treatment were collected, then the relevant PPI network was constructed and visualized by Cytoscape software (version 3.0; http://cytoscape.org/) and was further analyzed by ClusterONE, a plugin of Cytoscape. Given that most of biological networks were scale-free, the hub genes were then selected with a connectivity more than 4.

Function and pathway enrichment analysis

As a comprehensive set of functional annotation tools, the Database for Annotation, Visualization and Integrated Discovery (DAVID) has been used for the systematic and integrative analysis of large gene lists [19]. GO terms are significantly overrepresented in a set of genes from three aspects, including the cellular component, molecular function, and biological process [13, 20]. In this study, the significant GO terms and Kyoto Encyclopaedia of Genes and Genomes (KEGG, http://www.genome.jp/) pathway enrichment analyses of the identified overlapping DEGs were performed using DAVID with the thresholds of P-value < 0.05. Results were analyzed by using the IBM SPSS 19.0 software (International Business Machines Corporation, USA).

Network construction

A biological network was generated by the progesterone-mediated oocyte maturation pathway identified in KEGG. Based on preliminary relationships among differential genes, DEGs from the different treatment groups were assembled. The network analysis identified gene modules of the interconnected genes expressed co-ordinately in target pathways, followed by observation of genes that appeared to be involved in ovulation. The prediction of gene networks which were important to the ovulation process were validated using hCG- and RU486-stimulated experimental data (GSE54584). Details of the pathways, including constituent reactions, involved complexes, and network relationships, were elucidated in KEGG [20]. We recognized these pathways as elementary reactions that contained one reactant along with its resultant product to generate a gene network, regardless of small molecules.

Q-PCR verification

Sexually mature female rats (8-week-old, Sprague-Dawley) from Chongqing Medical University were used for the experimental verification. Rats were raised in controlled room conditions of constant temperature (22–25 °C) and 12 h light/dark cycles with adlibitum access to food and water. All experiments were performed according to the guidelines of the National Institutes of Health, and the experimental protocol was approved by the animal-experiment committee.

Seleceted genes were validated through analysis. There were two treatment groups (6 Sprague-Dawley female rats per group for one experiment). The first group was intraperitoneally injected with Pregnant Mare Serum Gonadotropin (PMSG, 20 IU), and then 48 h later, hCG (20 IU) was administered through intraperitoneal injection. The second group was based on the procedures of the first group. Specifically, the second group of rats was gavaged with RU486 (100 mg/kg), a progesterone receptor antagonist, at 37 h post-PMSG. And then, hCG was administered through intraperitoneal injection. Rats were sacrificed and cumulus oocyte complex (COCs) samples were collected after hCG treatment 12 h. Total RNA was acquired from ovary COCs using the extraction/isolation kit (BioTeke, Beijing, China). The RNA samples were reverse-transcribed into complementary DNA with the RevertAid First Strand cDNA Synthesis Kit (Takara, Dalian, China).

Each PCR reaction of q-PCR consisted 2 μl cDNA products, 7.5 μl of the Power SYBR Green PCR Master Mix (Takara, Dalian, China), 4.5 μl of water, and 1 μl of gene-specific primers. q-PCR was performed in the following conditions: activation at 95 °C for 30 s, followed by 40 cycles of 5 s at 95 °C and 30 s at 60 °C. The final melting curve was added to examine the amplification quality, whereas expression of mRNA for β-actin was viewed as an internal control standard. The sequences of the primers from National Center for Biotechnology Information reference sequences are shown in Table 1. The q-PCR results were analyzed using the Origin 8.0 software (Originlab Corporation, MA, US).

Table 1 Primer of prediction genes

Statistical analysis

The statistical significance of change in the gene expression was determined using the Student’s t-tests and posthoc Tukey’s test. The data were shown as a fold change (mean ± S.D) of values of at least three independent experiments and P < 0.05 was considered statistically significant.

Results

Functional and pathway analysis

To identify the genes and pathways involved in progestogenic networks, two data sets (GSE41836 and GSE54584, hCG treatment samples) were subjected to intercommunity analysis using bioinformatics approaches. Total 546 common DEGs were obtained from the overlapping differential genes (P < 0.05, absolute log2 FC > 1), which were then used for PPI analysis and visualization (Additional file 1: Figure S1). Results from ClusterONE analysis identified four hub genes, including PGR, reelin (RELN), phosphodiesterase 10A (PDE10A) and phospholipase A2 group IVA (PLA2G4A) (Additional file 2: Table S1).

In order to further determine the key genes in progestogenic networks, functional and pathway enrichment analyses of these four candidate genes showed that PGR was involved in the 51 functions mainly related to ovulation, such as regulation of the ovulation cycle and steroid hormone receptor activity, and participated the signaling pathway of progesterone regulation follicle maturation. RELN was involved in 47 different functions, such as the activity of protein kinases, and PDE10A was mainly reported in the 20 functions related to the nucleotide binding domain, particularly playing a role in the purine metabolic signaling pathway. Finally, PLA2G4A participated cell proliferation regulation, metabolism and other aspects of the functions, and mainly related to the MAPK signaling pathway and the GnRH signaling pathway (Table 2) [21, 22].

Table 2 Information of ovulation candidate genes

Furthermore, we selected the prominent functions of four genes shown in Table 3, including the significant biological process, cellular component, and molecular function related to hCG treatment. In addition, the top 25 statistical pathways were also exhibited as the pathway enrichment analysis results of the 546 DEGs, considering the involved gene numbers and P-vaule (Fig. 1).

Table 3 The prominent functions enrichment analysis including the molecular function, biological process, and cellular component
Fig. 1
figure 1

Pathway enrichment analysis. The top 25 statistics pathways identified by pathway enrichment analysis of the 546 DEGs, considering the involved gene numbers and P-vaule

Pathway regulation and gene network analysis

Progestogenic pathways are a coordinated process initiated by hormones that activate ovulation. PGR and RELN play an important role in this process. PGR is an upstream regulator of the progestogenic pathways in ovulation [23]. RELN acts as a factor promoting the end of follicle growth [24]. To verify whether the genes and networks of progestogenic pathways had potential theoretical significance, PGR and RELN were analyzed to investigate the effects of differential expression in this process.

To characterize genes’ alterations in preovulatory processes, gene expression data were analyzed related to hCG and RU486-treated experiments (GSE54584 data). Results demonstrated that significant changes in gene expression were observed comparing with the control groups. However, expression profiles of PGR in hCG and RU486-treated groups were significantly different. This transcript is known to be involved in progesterone-mediated oocyte maturation pathways and to interact with GNAI1, CPEB2 and KRAS (Fig. 2a). Four transcripts (MAPK1, CCNA2, IGF1 and BUB1) were significantly down-regulated in the two treatments (Fig. 2a and c). However, unlike the signaling pathways involving PGR, hCG and RU486-treated produced differential changes in RELN in follicular growth or focal adhesion pathways. Up-regulated transcripts in hCG-treated included PRKCA (> 5-fold), CAV1 (> 6-fold), EGFR (> 3-fold), RHOA (> 2-fold), ZYX (> 2-fold), VCL (> 4-fold), RAP1A (> 2-fold), and GRB2 (> 2-fold). In contrast, these transcripts were down-regulated in RU486-treated (Fig. 2b, d and e). In addition, some genes (MAPK1, BUB1, CANA2 and IGF1) involved in the RELN pathway were down-regulated in both hCG and RU486 treatment (Additional file 3: Table S2). These expression differences were considered relevant to the induction of ruptured and un-ruptured follicles and other features such as inflammation, tissue remodeling, extracellular matrix release, and steroid metabolism [2, 7, 17].

Fig. 2
figure 2

Gene expression analysis with hCG and RU treatment in preovulatory follicles from GSE54584. All validated genes were significantly differentially expressed in hCG- and RU486-treated ovarian. a and c were hCG-treated group, and b and d were RU486-treated group. Nodes in red represent the genes with expression level above the mean and nodes in green represent the genes with expression below the mean. The intensity of the pseudo-color reflects cross parts. e, Differentially expressed genes showed by visualized analysis with p-value < 0.05 and absolute log2 FC > 1

New gene network construction of the progestogenic pathway was based on primary network relationships between the four genes and pathways of PGR and RELN. The network combined 8 pathways (progesterone-mediated oocyte maturation, MAPK signaling pathway, GnRH signaling pathway, purine metabolism, PI3K-Akt signaling pathway, Wnt signaling pathway, ECM-receptor interaction, and focal adhesion) with node genes PIK3R5, MAPK, and ADCY1, then added nine predicted genes (GNAI1, PRKCA, RHOA, ZYX, VCL, RAP1A, CAV1, EGFR and GRB2) to the network. A combined method was taken, regardless of small molecules or undetected genes (Fig. 3).

Fig. 3
figure 3

Visualized progesterone-related genes network. Nodes in different colors represent different target genes or pathways. Purple sexangles represent node genes (MAPK, SRC, PTK2, ITGA, CAPN2, GPCR, ADCY1) and predicted target genes (GNAI1, PRKCA, CAV1, EGFR, RHOA, ZYX, VCL, GRB2 and RAP1A) in this study; yellow color ellipses represent genes between networks; and red color rectangles reflect pathways involved in progestogenic networks

Validation of predicted results by q-PCR

In order to verify whether the differential genes (GNAI1, PRKCA, RHOA, ZYX, VCL, RAP1A, CAV1, EGFR and GRB2) in the progesterone signaling pathway were indeed regulated by PGR, they were detected by q-PCR. This was the quantitative detection of samples from hCG- and RU486-treated rats after the extraction of COCs. The results of q-PCR showed that the expression of PGR was almost completely inhibited after RU486 treatment, and the expression of RELN was also decreased (Fig. 4a and b). In addition, it was found that the nine predicted genes (GNAI1, PRKCA, RHOA, ZYX, VCL, RAP1A, CAV1, EGFR and GRB2) exhibited a differential change (Fig. 4c-k). The expression of VCL, RHOA, RAP1A and GRB2 decreased by about 3-fold compared with the control group, and the significant decreases of ZYX, GNAI1, CAV1 and PRKCA were about 2-fold. The change of EGFR was inhibited, although the trend became below par. This may also be subject to the regulation of other factors in the process of ovulation.

Fig. 4
figure 4

Validation of progesterone-related predicted genes by q-PCR analysis. The two groups of rats were treated by PMSG and hCG or PMSG, hCG and RU486. Total RNA of COCs was extracted for q-PCR analysis of the genes expression. a-b: PGR and RELN expression; c-k: expression of the nine genes (GNAI1, PRKCA, CAV1, EGFR, RHOA, ZYX, VCL, GRB2 and RAP1A, respectively). Different asterisks indicate statistically significant differences (*, P < 0.05; **, P < 0.01). Error bars represent S.D.

Discussion

One of the challenges in mammalian ovulation research is to investigate the basic regulatory and physiological mechanisms [25]. The present study has identified genes with special importance in unique developmental processes. Selectively GSE41836 and GSE54584 data served as accessible data sets for studying the progestogenic-related gene networks in mammals [10]. Herein, we highlighted ovarian physiology and identified many new progestogenic-related genes in this study.

This study also identified several novel initiation factors of ovulation. Regulations of hormones and steroidogenesis are regarded as an initiation process, which provides detailed fundamental information on the profound changes regulated by gene networks in ovulation. Hormone (FSH and LH) induced primary follicle maturation and eventually approached the pre-ovulatory stage in mice and rats [2, 26]. Pituitary-released LH forms the LH surge which subsequently initiates the estrous cycle in rodents in response to the high levels of estrogen [27]. The steroid hormone progesterone, regulated by the LH surge, plays a key role in ovulation [28]. The effects of progesterone were mediated by PGR, and its expression was observed in the granulosa cells of pre-ovulatory follicles following hCG treatment in mice and rats [29]. PGR was accompanied by the coordinated expression of progesterone biosynthetic enzymes, including steroidogenic acute regulatory protein (Star) and cholesterol side-chain cleavage enzyme (Cyp11a1) [30, 31]. An expression increase of Star and Cyp11a1 was observed following hCG stimulation, but a decrease was in RU486-treated mice, which revealed a pattern of expression related to the progestogenic pathway.

In the progestogenic pathway, PGR expression is induced primarily in granulosa cells of the pre-ovulatory follicles [2]. PGR is a transcription factor involved in multiple processes including oocyte maturation, ovulation, ovulation cycle process, epithelial cell development, and steroid hormone receptor signaling pathways. PGR directly mediates ovulation and indirectly regulates other ovulation-related factors. The expression of RELN, a regulator of follicle growth factor like IGF2 [24], decreased in hCG-treated rats (Fig. 2c). It binded with the receptor LRP8 to regulate follicular growth in cattle. MAPK8IP1 encoding LRP8 intracellular interacting partner was also expressed in granulosa cells of the dominant follicle. This differential expression pattern suggested an RELN/LRP8/MAPK8IP1 paracrine interaction to regulate follicular growth until ovulation [32]. PDE10A, one of the PDE families, is also expressed in bovine cumulus cells by cAMP-PDE activity [33]. Mammalian oocytes initiate meiosis in the foetus but remain arrested at the diplotene stage of the first meiotic prophase, before being potentiality to resume by the ovulatory luteinising hormone pulse [34]. High levels of cAMP keep oocytes in the meiotic arrest phase by activating cAMP-dependent protein kinase. As for PDE10A, effects of ovulation on the process are uncertain. Functional and pathway analysis found that PLA2G4A was involved in the MAPK and GnRH signaling pathways. Many studies have confirmed that ovulation is associated with an LH/hCG-dependent induction of PLA2G4A in granulosa cells via the adenylyl cyclase/cAMP pathway [35]. Kurusu et al. [36] found that PLA2G4A optimizes ovulation and fertilization when coupled with prostaglandin endoperoxide synthase in female mice lacking PLA2G4A. This study found that four genes (PGR, RELN, PDE10A, and PLA2G4A) developed an expression pattern consistent with a progestogenic pathway during ovulation in mammals. This analysis suggests that these four genes can form a network of cellular pathways or nodes to regulate ovulation.

Progestogenic pathway plays an important role in the “story” of follicle maturation and ovulation. In order to explore extensions of the progestogenic pathway, this study investigated the four genes that were involved in this pathway. An analysis of the four genes from the KEGG pathway and PPI showed that MAPK, PIK3R5, PTK2, ITGA, and ADCY1 had the most regulatory connections to adjacent genes (Fig. 4). These genes are considered important in regulating features involved in the progestogenic network or as downstream factors of the network. Genes involved in each pathway may act in concert with accomplishing distinct processes during ovulation including prostaglandin synthesis (Ptgs2 and PLA2G4A) [7], cholesterol uptake (Ldlr and Scarb1) [37], regulation of progesterone synthesis (Star) [30], inactivation of estrogen (Sult1e1) [38], and downstream effectors of LH signaling (Pgr, Cebpb, Areg, Ereg, and Adamts1) [6, 26, 29, 39]. These processes regulate inflammation, follicle-wall degradation, hormone balance and the induction of certain paracrine factors. In addition, genes encode proteins involved in various biology process, including reproductive developmental processes and granulosa cell expansion (Tgfb2, Foxl2, Pde3a, Esr2, Fshr, Ccnd2) [40], amino acid degradation and ketogenesis (Hmgcs2, Cpt1b) [41]. These results presented candidate genes involved in independent cellular pathways to carry out relevant functions in progestogenic pathways.

Interestingly, when the rats were subjected to hCG and RU486 treatment, the results indicated that most of the genes involved in PGR and RELN pathways were differentially regulated. Furthermore, these results illustrated that focal adhesion, EMC-receptor interaction and MAPK signaling pathways were affected in hCG- and RU486-treated rats. MAPK signaling pathways provoke ovulation and collapse of the follicle, which were stimulated by a variety of exotic signals (MAP kinases 1 and 3, extracellular-regulated protein kinases 1 and 2 (ERK1/2)) [42]; the Wnt-signaling and PI3K-Akt signaling pathways were not significantly affected by hCG treatment (Fig. 2c). Conti and Makker [43, 44] found that the PI3K/PTEN/Akt signaling pathway and cAMP/ERK/EGF network induced oocyte and meiotic maturation. It is noteworthy that the PDE family participated in the cAMP pathway and research suggests that it also regulates ovulation. Sasseville et al. [33] indicated the establishment of a model of cell-specific expression of PDEs in the ovarian follicle. This suggested that Pde3a was a major isoform of oocyte PDE, Pde4d was the major granulosa cell PDE isoforms and PDE10A was expressed in cumulus cells. Cell-specific expression of cAMP-PDEs in ovarian follicles has implications for understanding the hormonal regulation of folliculogenesis [45]. Further studies are needed to determine how Pde10a-Adcy1 participates in progestogenic pathways. Pathway analysis and gene validation indicated that RELN could participate in PI3K-Akt signaling by PTEN, but the molecular mechanism of this process remained unclear. However, when RELN combines with receptor LRP8 to regulate follicular growth, the pattern of LRP8 expression correlates with the capacity of follicles to synthesize higher steroid levels in cattle [32]. Subsequently, the phase of follicular development, including the LH surge, changes the relationship between progestin and androgen. Enzymes required for progestin include hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase (Hsd3b2), and Cyp11a1. This suggests that RELN and LRP8 may be regulated in progestogenic pathways, but human studies have not verified this regulation. Pathway validation indicated the PRKCA exhibited significant changes in RELN regulation (Fig. 2b and d). This validation also connected Ptk2 to progesterone-mediated oocyte maturation which confirmed the relationship between RELN and PGR. Details of this interaction require further investigation. Relative expression results using q-PCR also found that GNAI1 was considered as a PGR indirect regulatory factor, but this process was not previously associated with ovulation. GNAI1, PRKCA, RHOA, ZYX, VCL, RAP1A, CAV1, EGFR and GRB2 produced changes in progestogenic pathways and these factors were considered as ovulation-associated factors. PLA2G4A is involved in the progestogenic pathway by paracrine regulation, which plays an important role in ovulation. The ovulatory surge of LH regulates the expression of each component of the PGE2 synthesis-metabolism-transport pathway to mediate key ovulatory events including cumulus expansion, rupture and release of oocytes [7].

Conclusions

In sum, a systematic bioinformatics analysis approach could provide an overview of a particular developmental process. Results suggested that the four genes (PGR, RELN, PDE10A and PLA2G4A) were involved in novel progestogenic networks (Fig. 3). These networks provided important regulatory genes and signaling pathways which were involved in ovulation. At the same time, hCG- and RU486-treated experiments confirmed that candidate genes (GNAI1, PRKCA, RHOA, ZYX, VCL, RAP1A, CAV1, EGFR and GRB2) were involved in ovulation.

This study has shown that the above genes are selectively or highly differentially expressed in progestogenic pathways, which provides important information for future downstream analysis to identify the significant genes in ovulation and develop effective strategies for fertility control.

Abbreviations

Cyp11a1:

Cholesterol side-chain cleavage enzyme

E2:

Estradiol

ECG:

Equine chorionic gonadotrophin

FSH:

Follicle-stimulating hormone

GnRH:

Gonadotropin-releasing hormone

GO:

Gene Ontology

HCG:

Human chorionic gonadotrophin

Hsd3b2:

Hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase

Il6:

Interleukin 6

LH:

Luteinizing hormone

OCC:

Oocyte cumulus complex

PDE10A:

Phosphodiesterase 10A

PGR:

Progesterone receptor

PLA2G4A:

Phospholipase A2 group IVA

Ptgs2:

Prostaglandin-endoperoxide synthase 2

RELN:

Reelin

Vegfa:

Vascular endothelial growth factor

References

  1. Deady LD, Sun JA. Follicle rupture assay reveals an essential role for follicular adrenergic signaling in drosophila ovulation. PLoS Genet. 2015;11(10):e1005604.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kim J, Bagchi IC, Bagchi MK. Control of ovulation in mice by progesterone receptor-regulated gene network. Mol Hum Reprod. 2009;15(12):821–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kadariya I, Wang J, ur Rehman Z, Ali H, Riaz H, He J, Bhattarai D, Liu JJ, Zhang SJ. RNAi-mediated knockdown of inhibin α subunit increased apoptosis in granulosa cells and decreased fertility in mice. J Steroid Biochem Mol Biol. 2015;152:161–70.

    Article  CAS  PubMed  Google Scholar 

  4. Cho J, Kim H, Kang DW, Yanagisawa M, Ko C. Endothelin B receptor is not required but necessary for finite regulation of ovulation. Life Sci. 2012;91(13–14):613–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burns KH, Yan C, Kumar TR, Matzuk MM. Analysis of ovarian gene expression in follicle-stimulating hormone beta knockout mice. Endocrinology. 2001;142(7):2742–51.

    Article  CAS  PubMed  Google Scholar 

  6. Blaha M, Nemcova L, Kepkova KV, Vodicka P, Prochazka R. Gene expression analysis of pig cumulus-oocyte complexes stimulated in vitro with follicle stimulating hormone or epidermal growth factor-like peptides. Reporod Biol Endocrinol. 2015;13:113.

    Article  Google Scholar 

  7. Duffy DM. Novel contraceptive targets to inhibit ovulation: the prostaglandin E2 pathway. Hum Reprod Update. 2015;21(5):652–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mauro A, Martelli A, Berardinelli P, Russo V, Bernabò N, Di Giacinto O, Mattioli M, Barboni B. Effect of antiprogesterone RU486 on VEGF expression and blood vessel remodeling on ovarian follicles beforeovulation. PLoS One. 2014;9(4):e95910.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Molyneaux KA, Schaible K, Wylie C. GP130, the shared receptor for the LIF/IL6 cytokine family in the mouse, is not required for early germ cell differentiation, but is required cell-autonomously in oocytes for ovulation. Development. 2003;130(18):4287–94.

    Article  CAS  PubMed  Google Scholar 

  10. Fan HY, Liu Z, Shimada M, Sterneck E, Johnson PF, Hedrick SM, Richards JS. MAPK3/1 (ERK1/2) in ovarian granulosa cells are essential for female fertility. Science. 2009;324(5929):938–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang JY, Wu Y, Zhao S, Liu ZX, Zeng SM, Zhang GX. Lysosomes are involved in induction of steroidogenic acute regulatory protein (StAR) gene expression andprogesterone synthesis through low-density lipoprotein in cultured bovine granulosa cells. Theriogenology. 2015;84(5):811–7.

    Article  CAS  PubMed  Google Scholar 

  12. Zhou C, Teng WJ, Zhuang J, Liu HL, Tang SF, Cao XJ, Qin BN, Wang CC, Sun CG. Analysis of the gene-protein interaction network in glioma. Genet Mol Res. 2015;14(4):14196–206.

    Article  CAS  PubMed  Google Scholar 

  13. Wang Y, Huang L, Wu S, Jia Y, Yang Y, Luo L, Bi A, Fang M. Bioinformatics analyses of the role of vascular endothelial growth factor in patients with non-small cell lung Cancer. PLoS One. 2015;10(9):e0139285.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Nilsson E, Zhang B, Skinner MK. Gene bionetworks that regulate ovarian primordial follicle assembly. BMC Genomics. 2013;14:496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li C, Zhu B, Chen J, Huang X. Novel prognostic genes of diffuse large B-cell lymphoma revealed by survival analysis of gene expression data. Onco Targets Ther. 2015;8:3407–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Grøndahl ML, Borup R, Vikeså J, Ernst E, Andersen CY, Lykke-Hartmann K. The dormant and the fully competent oocyte: comparing the transcriptome of human oocytes from primordial follicles and in metaphase II. Mol Hum Reprod. 2013;19(9):600–17.

    Article  PubMed  Google Scholar 

  17. Tsubota K, Kanki M, Noto T, Nakatsuji S, Oishi Y, Matsumoto M, Nakayama H. Altered gene expression profile in ovarian follicle in rats treated with indomethacin and RU486. J Toxicol Sci. 2015;40(3):413–25.

    Article  CAS  PubMed  Google Scholar 

  18. Agca C, Yakan A, Agca Y. Estrus synchronization and ovarian hyper-stimulation treatments have negligible effects on cumulus oocytecomplex gene expression whereas induction of ovulation causes major expression changes. Mol Reprod Dev. 2013;80(2):102–17.

    Article  CAS  PubMed  Google Scholar 

  19. Kutmon M, Evelo CT, Coort SLA. Network biology workflow to study transcriptomics data of the diabetic liver. BMC Genomics. 2014;15:971.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kou Y, Zhang S, Chen X, Hu S. Gene expression profile analysis of colorectal cancer to investigate potential mechanisms using bioinformatics. Onco Targets Ther. 2015;8:745–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Wismann J, Willoughby D. Gender differences in carbohydrate metabolism and carbohydrate loading. J Int Soc Sports Nutr. 2006;3:28–34.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhang W, Zhou T. A sparse reconstruction approach for identifying gene regulatory networks using steady-state experiment data. PLoS One. 2015;10(7):e0130979.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Wissing ML, Kristensen SG, Andersen CY, Mikkelsen AL, Høst T, Borup R, Grøndahl ML. Identification of new ovulation-related genes in humans by comparing the transcriptome of granulosa cells before and after ovulation triggering in the same controlled ovarian stimulation cycle. Hum Reprod. 2014;29(5):997–1010.

    Article  CAS  PubMed  Google Scholar 

  24. Nivet AL, Vigneault C, Blondin P, Sirard MA. Changes in granulosa cells' gene expression associated with increased oocyte competence in bovine. Reproduction. 2013;145(6):555–65.

    Article  CAS  PubMed  Google Scholar 

  25. Cerri RL, Thompson IM, Kim IH, Ealy AD, Hansen PJ, Staples CR, Li JL, Santos JE, Thatcher WW. Effects of lactation and pregnancy on gene expression of endometrium of Holstein cows at day 17 of the estrous cycle or pregnancy. J Dairy Sci. 2012;95(10):5657–75.

    Article  CAS  PubMed  Google Scholar 

  26. Bylander A, Gunnarsson L, Shao R, Billig H, Larsson DG. Progesterone-mediated effects on gene expression and oocyte-cumulus complex transport in the mouse fallopiantube. Reprod Biol Endocrinol. 2015;13:40.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Breen SM, Andric N, Ping T, Xie F, Offermans S, Gossen JA, Ascoli M. Ovulation involves the luteinizing hormone-dependent activation of Gq/11 in granulosa cells. Mol Endocrinlo. 2013;27(9):1483–91.

    Article  CAS  Google Scholar 

  28. Tam KK, Russell DL, Peet DJ, Bracken CP, Rodgers RJ, Thompson JG, Kind KL. Hormonally regulated follicle differentiation and luteinization in the mouse is associated with hypoxia inducible factor activity. Mol Cell Endocrinol. 2010;327(1–2):47–55.

    Article  CAS  PubMed  Google Scholar 

  29. Robker RL, Russell DL, Espey LL, Lydon JP, Malley BW, Richards JS. Progesterone-regulated genes in the ovulation process: ADAMTS-1 and cathepsin L proteases. Proc Natl Acad Sci U S A. 2000;97(9):4689–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kowalewski MP, Gram A, Boos A. The role of hypoxia and HIF1α in the regulation of STAR-mediated steroidogenesis in granulosa cells. Mol Cell Endocrinol. 2015;401:35–44.

    Article  CAS  PubMed  Google Scholar 

  31. Cui J, Shen Y, Li R. Estrogen synthesis and signaling pathways during ageing: from periphery to brain. Trends Mol Med. 2013;19(3):197–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Fayad T, Lefebvre R, Nimpf J, Silversides DW, Lussier JG. Low-density lipoprotein receptor-related protein 8 (LRP8) is upregulated in granulosa cells of bovine dominantfollicle: molecular characterization and spatio-temporal expression studies. Biol Reprod. 2007;76(6):466–75.

    Article  CAS  PubMed  Google Scholar 

  33. Sasseville M, Albuz FK, Côté N, Guillemette C, Gilchrist RB, Richard FJ. Characterization of novel phosphodiesterases in the bovine ovarian follicle. Biol Reprod. 2009;81(2):415–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Thomas RE, Thompson JG, Armstrong DT, Gilchrist RB. Effect of specific phosphodiesterase isoenzyme inhibitors during in vitro maturation of bovine oocytes on meiotic and developmental capacity. Biol Reprod. 2004;71(4):1142–9.

    Article  CAS  PubMed  Google Scholar 

  35. Diouf MN, Sayasith K, Lefebvre R, Silversides DW, Sirois J, Lussier JG. Expression of phospholipase A2 group IVA (PLA2G4A) is upregulated by human chorionic gonadotropin in bovine granulosa cells of ovulatory follicles. Biol Reprod. 2006;74(6):1096–103.

    Article  CAS  PubMed  Google Scholar 

  36. Kurusu S, Sapirstein A, Bonventre JV. Group IVA phospholipase A2 optimizes ovulation and fertilization in rodents through induction of and metaboliccoupling with prostaglandin endoperoxide synthase 2. FASEB J. 2012;26:3800–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Park JY, Jang H, Curry TE, Sakamoto A, Jo M. Prostate androgen-regulated mucin-like protein 1: a novel regulator of progesterone metabolism. Mol Endocrinol. 2013;27(11):1871–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gerrits H, Paradé MC, Koonen-Reemst AM, Bakker NE, Timmer-Hellings L, Sollewijn Gelpke MD, Gossen JA. Reversible infertility in a liver receptor homologue-1 (LRH-1)-knockdown mouse model. Reprod Fertil. 2014;26(2):293–306.

    Article  CAS  Google Scholar 

  39. Fan HY, Liu Z, Johnson PF, Richards JS. CCAAT/enhancer-binding proteins(C/EBP)-α and -β are essential for ovulation, luteinization, and the expression of key target genes. Mol Endocrinol. 2011;25:253–68.

    Article  CAS  PubMed  Google Scholar 

  40. Regan SL, McFarlane JR, O'Shea T, Andronicos N, Arfuso F, Dharmarajan A, Almahbobi G. Flow cytometric analysis of FSHR, BMRR1B, LHR and apoptosis in granulosa cells and ovulation rate in merino sheep. Reproduction. 2015;150(2):151–63.

    Article  CAS  PubMed  Google Scholar 

  41. Dunning KR, Cashman K, Russell DL, Thompson JG, Norman RJ, Robker RL. Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development. Biol Reprod. 2010;83:909–18.

    Article  CAS  PubMed  Google Scholar 

  42. Duggavathi R, Murphy BD. Development. Ovulation signals. Science. 2009;324(5929):890–1.

    Article  CAS  PubMed  Google Scholar 

  43. Conti M, Hsieh M, Zamah AM, Oh JS. Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Mol Cell Endocrinol. 2012;356(1–2):65–73.

    Article  CAS  PubMed  Google Scholar 

  44. Makker A, Goel MM, Mahdi AA. PI3K/PTEN/Akt and TSC/mTOR signaling pathways, ovarian dysfunction, and infertility: an update. J Mol Endocrinol. 2014;53:R103–18.

    Article  CAS  PubMed  Google Scholar 

  45. Campos-Chillon F, Farmerie TA, Bouma GJ, Clay CM, Carnevale EM. Effects of aging on gene expression and mitochondrial DNA in the equine oocyte and follicle cells. Reprod Fertil Dev. 2015;27(6):925–33.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We gratefully thank Dr. Muhammad Farrukh Nisar, Dr. Feng Jianguo and Dr. Kong Xiangjun for grammar revision.

Funding

This work was supported by National Natural Science Foundation of China (Grant No. 31372287), National Biological Breeding Capacity Building and Industrialization Projects (2014–2573), Ministry of Agriculture transgenic major projects of China (2014ZX0800952B), and the Agricultural Science and Technology Innovation Program of China (ASTIP-IAS13).

Availability of data and materials

The data supporting the results of this article are included and cited within the article and its additional files.

Author information

Authors and Affiliations

Authors

Contributions

FY carried out the molecular genetic studies, completed in the sequence alignment and drafted the manuscript. MW helped to finish the sequence analysis. FY and PW designed this study and performed the statistical analysis. BZ, MC, WX and KZ participated in its design and coordination, and provided valuable advice. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Pingqing Wang.

Ethics declarations

Ethics approval and consent to participate

This study does not contain any studies with human participants or animals performed by any of the authors. Informed consent was obtained from all individual participants included in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Additional files

Additional file 1:

Figure S1. The significant modules in the protein-protein interaction network with MCODE. Nodes denoted proteins (genes). In specific, the yellow denoted seed proteins; the red denoted the up-regulated proteins; and the green denoted down-regulated proteins; the width of edges was determined according to the combined score of the protein-protein interaction relationships. Abbreviation: MCODE, Molecular Complex Detection. (DOCX 115 kb)

Additional file 2:

Table S1. Four genes information from analysis of Cluster One. (DOCX 13 kb)

Additional file 3:

Table S2. The list of significantly differentially expressed genes of the pathways involving RELN and PGR. (DOCX 14 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, F., Wang, M., Zhang, B. et al. Identification of new progestogen-associated networks in mammalian ovulation using bioinformatics. BMC Syst Biol 12, 36 (2018). https://0-doi-org.brum.beds.ac.uk/10.1186/s12918-018-0577-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12918-018-0577-7

Keywords