Skip to main content

Identification of key pathways and genes in polycystic ovary syndrome via integrated bioinformatics analysis and prediction of small therapeutic molecules

Abstract

To enhance understanding of polycystic ovary syndrome (PCOS) at the molecular level; this investigation intends to examine the genes and pathways associated with PCOS by using an integrated bioinformatics analysis. Based on the expression profiling by high throughput sequencing data GSE84958 derived from the Gene Expression Omnibus (GEO) database, the differentially expressed genes (DEGs) between PCOS samples and normal controls were identified. We performed a functional enrichment analysis. A protein-protein interaction (PPI) network, miRNA- target genes and TF - target gene networks, were constructed and visualized, with which the hub gene nodes were identified. Validation of hub genes was performed by using receiver operating characteristic (ROC) and RT-PCR. Small drug molecules were predicted by using molecular docking. A total of 739 DEGs were identified, of which 360 genes were up regulated and 379 genes were down regulated. GO enrichment analysis revealed that up regulated genes were mainly involved in peptide metabolic process, organelle envelope and RNA binding and the down regulated genes were significantly enriched in plasma membrane bounded cell projection organization, neuron projection and DNA-binding transcription factor activity, RNA polymerase II-specific. REACTOME pathway enrichment analysis revealed that the up regulated genes were mainly enriched in translation and respiratory electron transport and the down regulated genes were mainly enriched in generic transcription pathway and transmembrane transport of small molecules. The top 10 hub genes (SAA1, ADCY6, POLR2K, RPS15, RPS15A, CTNND1, ESR1, NEDD4L, KNTC1 and NGFR) were identified from PPI network, miRNA - target gene network and TF - target gene network. The modules analysis showed that genes in modules were mainly associated with the transport of respiratory electrons and signaling NGF, respectively. We find a series of crucial genes along with the pathways that were most closely related with PCOS initiation and advancement. Our investigations provide a more detailed molecular mechanism for the progression of PCOS, detail information on the potential biomarkers and therapeutic targets.

Introduction

Polycystic ovary syndrome (PCOS) is one of the most prevalent endocrine disorder around the world, with an estimated about one in 15 women worldwide [1]. PCOS exposes patients to a major psychosocial burden and is characterized by hyperandrogenism and chronic anovulation [2]. Diabetes, heart disease, obesity, non-alcoholic fatty liver disease and hypertension are the risk factors associated with PCOS [3,4,5,6,7]. Therefore, it is of prime importance to identify the etiological factors, molecular mechanisms, and pathways to discover novel diagnostic markers, prognostic markers and therapeutic targets for PCOS.

Numerous research strategies have recently investigated the molecular mechanisms of PCOS. High-throughput RNA sequencing technology has received extensive attention among these research strategies and has generated significant advances in the field of endocrine disorder with marked clinical applications ranging from molecular diagnosis to molecular classification, patient stratification to prognosis prediction, and discovery of new drug targets to response prediction [8]. In addition, gene expression profiling investigation on PCOS have been performed using high-throughput RNA sequencing, and several key genes and diagnostic biomarkers have been diagnosed for this syndrome, including the profiling of many of differentially expressed genes (DEGs) associated in different pathways, biological processes, or molecular functions [9]. Integrated bioinformatics analyses of expression profiling by high throughput sequencing data derived from different investigation of PCOS could help identify the novel diagnostic markers, prognostic markers and further demonstrate their related functions and potential therapeutic targets in PCOS.

Therefore, in the current investigation, the dataset (GSE84958) was then retrieved from the publicly available Gene Expression Omnibus database (GEO, http://www.ncbi.nlm.nih.gov/geo/) [10] to identify DEGs and the associated biological processes PCOS using comprehensive bioinformatics analyses. The DEGs were subjected to functional enrichment and pathway analyses; moreover, a protein-protein interaction (PPI) network, miRNAs - target gene regulatory network and TFs - target gene regulatory network were constructed to screen for key genes, miRNA and TFs. The aim of this investigation was to identify key genes and pathways in PCOS using bioinformatics analysis, and then to explore the molecular mechanisms of PCOS and categorize new potential diagnostic therapeutic biomarkers of PCOS. We anticipated that these investigations will provide further understanding of PCOS pathogenesis and advancement at the molecular level.

Materials and Methods

RNA sequencing data

Expression profiling by high throughput sequencing dataset GSE84958 was downloaded from NCBI-GEO, a public database of next-generation sequencing, to filter the DEGs between PCOS and normal control. The expression profiling by high throughput sequencing GSE84958 was based on GPL16791 platforms (Illumina HiSeq 2500 (Homo sapiens)) and consisted of 30 PCOS samples and 23 normal control.

Identification of DEGs

The limma [11] in R bioconductor package was used to analyze the DEGs between PCOS samples and normal control samples in the expression profiling by high throughput sequencing data of GSE84958. The adjusted P-value and [logFC] were calculated. The Benjamini & Hochberg false discovery rate method was used as a correction factor for the adjusted P-value in limma [12]. The statistically significant DEGs were identified according to P<0.05, and [logFC] > 2.5 for up regulated genes and [logFC] < -1.5 for down regulated genes. All results of DEGs were downloaded in text format, hierarchical clustering analysis being conducted.

GO and pathway enrichment of DEGs in PCOS

To reflect gene functions, GO (http://geneontology.org/) [13] has been used in three terms: biological processes (BP), cellular component (CC) and molecular function (MF). ToppGene (ToppFun) (https://toppgene.cchmc.org/enrichment.jsp) [14] is an online database offering a comprehensive collection of resources for functional annotation to recognize the biological significance behind a broad list of genes. The functional enrichment analyses of DEGs, including GO analysis and REACTOME (https://reactome.org/) [15] pathway enrichment analysis, were performed using ToppGene in the present study, using the cut-off criterion P-value<0.05 and gene enrichment count>2.

PPI networks construction and module analysis

The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING: http://string-db.org/) is online biological database and website designed to evaluate PPI information [16] Proteins associated with DEGs were selected based on information in the STRING database (PPI score >0.7), and then PPI networks were constructed using Cytoscape software (http://cytoscape.org/) [17]. In this investigation, node degree [18], betweenness centrality [19], stress centrality [20] and closeness centrality [21], these constitutes a fundamental parameters in network theory, were adopted to calculate the nodes in a network. The topological properties of hub genes were calculated using Cytoscape plugin Network Analyzer. The PEWCC1 (http://apps.cytoscape.org/apps/PEWCC1) [22], a plugin for Cytoscape, was used to screen the modules of the PPI network. The criteria were set as follows: degree cutoff=2, node score cutoff=0.2, k-core=2 and maximum depth=100. Moreover, the GO and pathway enrichment analysis were performed for DEGs in these modules.

Construction of miRNA - target regulatory network

Furthermore, the target genes of the significant target genes were predicted by using miRNet database (https://www.mirnet.ca/) [23], when the miRNAs shared a common target gene. Finally, the miRNA - target genes regulatory network depicted interactions between miRNAs and their potential targets in PCOS were visualized by using Cytoscape.

Construction of TF - target regulatory network

Furthermore, the target genes of the significant target genes were predicted by using TF database (https://www.mirnet.ca/) [23], when the TFs shared common target genes. Finally, the TF- target genes regulatory network depicted interactions between TFs and their potential targets in PCOS were visualized by using Cytoscape.

Receiver operating characteristic (ROC) curve analysis

The ROC curve was used to evaluate classifiers in bioinformatics applications. To further assess the predictive accuracy of the hub genes, ROC analysis was performed to discriminate PCOS from normal control. ROC curves for hub genes were generated using pROC in R [24] based on the obtained hub genes and their expression profiling by high throughput sequencing data. The area under the ROC curve (AUC) was determined and used to compare the diagnostic value of hub genes.

Validation of the expression levels of candidate genes by RT-PCR

Total RNA was extracted from PCOS (UWB1.289 (ATCC® CRL-2945™)) and normal ovarian cell line (MES-OV (ATCC® CRL-3272™)) using TRI Reagent® (Sigma, USA). The Reverse transcription cDNA kit (Thermo Fisher Scientific, Waltham, MA, USA) and 7 Flex real-time PCR system (Thermo Fisher Scientific, Waltham, MA, USA) were used for reverse transcription and real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) assay. Polymerase chain reaction primer sequences are listed in Table 1. β-actin was used as an internal control for quantification. The relative expression levels of target transcripts were calculated using the 2-∆∆Ct method [25]. The thermocycling conditions used for RT-PCR were as follows: initial denaturation at 95°C for 15 min, followed by 40 cycles at 95°C for 10 sec, 60°C for 20 sec and 72°C for 20 sec.

Table 1 Primers used for quantitative PCR

Molecular docking studies

Surflex-docking studies of the standard drug molecule used in polycystic ovary syndrome were used on over expressed genes and were collected from PDB data bank using perpetual SYBYL-X 2.0 software. Using ChemDraw Software, all the drug molecules were illustrated, imported and saved in sdf. templet using open babel free software. The protein structures of POLR2K (), RPS15, RPS15 alpha and SAA1 of their co-crystallised protein of PDB code 1LE9, 3OW2, 1G1X and 4IP8 respectively were extracted from Protein Data Bank [26,27,28]. Gasteiger Huckel (GH) charges were applied along with the TRIPOS force field to all the drug molecules and is standard for the structure optimization process. In addition, energy minimization was achieved using MMFF94s and MMFF94 algorithm methods. The protein preparation was carried out after incorporation of protein. The co-crystallized ligand was extracted from the crystal structure and all water molecules; more hydrogen was added and the side chain was set. For energy minimisation, the TRIPOS force field was used. The interaction efficiency of the compounds with the receptor was represented in kcal / mol units by the Surflex-Dock score. The interaction between the protein and the ligand, the best pose was incorporated into the molecular area. The visualization of ligand interaction with receptor is done by using discovery studio visualizer.

Results

Identification of DEGs

Expression profiling by high throughput sequencing dataset was obtained from the National Center for Biotechnology Information GEO database containing PCOS samples and normal control samples: GSE84958. Then, the R package named “limma” was processed for analysis with adjusted P < 0.05, and [logFC] > 2.5 for up regulated genes and [logFC] < -1.5 for down regulated genes. All DEGs were displayed in volcano maps (Fig. 1). A total of 739 DEGs including 360 up regulated and 379 down regulated genes (Table 2) were identified in PCOS samples compared to normal control samples. The results are shown in the heatmap (Fig. 2).

Fig. 1
figure 1

Volcano plot of differentially expressed genes. Genes with a significant change of more than two-fold were selected. Green dot represented up regulated significant genes and red dot represented down regulated significant genes

Table 2 The statistical metrics for key differentially expressed genes (DEGs)
Fig. 2
figure 2

Heat map of differentially expressed genes. Legend on the top left indicate log fold change of genes. (A1 – A2 = normal control samples; B1 – B30 = PCOS samples)

GO and pathway enrichment of DEGs in PCOS

The top 739 DEGs were chosen to perform GO and REACTOME pathway enrichment analyses. Gene Ontology (GO) analysis identified that the DEGs were significantly enriched in BP, including the peptide metabolic process, intracellular protein transport, plasma membrane bounded cell projection organization and cell morphogenesis (Table 3). In terms of CC, DEGs were mainly enriched in organelle envelope, catalytic complex, neuron projection and cell junction were the most significantly enriched GO term (Table 3). In addition, MF demonstrated that the DEGs were enriched in the RNA binding, transcription factor binding, DNA-binding transcription factor activity, RNA polymerase II-specific and ATP binding (Table 3). REACTOME pathway enrichment analysis was used to screen the signaling pathways for differential genes. These DEGs were mainly involved in the translation, respiratory electron transport, generic transcription pathway and transmembrane transport of small molecules (Table 4).

Table 3 The enriched GO terms of the up and down regulated differentially expressed genes
Table 4 The enriched pathway terms of the up and down regulated differentially expressed genes

PPI networks construction and module Analysis

Following the analysis based on the PPI networks, 4141 nodes and 14853 edges were identified in Cytoscape (Fig. 3a). The genes with higher scores were the hub genes, as the genes of node degree, betweenness centrality, stress centrality, closeness centrality may be linked with PCOS. The top 10 hub genes were SAA1, ADCY6, POLR2K, RPS15, RPS15A, ESR1, LCK, S1PR5, CCL28 and CTNND1 and are listed in Table 5. Enrichment analysis demonstrated that module 1 (Fig. 3b) and module 2 (Fig. 3c) might be associated with respiratory electron transport, organelle envelope, catalytic complex, gene expression, signaling by NGF and neuron projection.

Fig. 3
figure 3

PPI network and the most significant modules of DEGs. a The PPI network of DEGs was constructed using Cytoscape. b The most significant module was obtained from PPI network with 26 nodes and 160 edges for up regulated genes. c The most significant module was obtained from PPI network with 26 nodes and 71 edges for up regulated genes. Up regulated genes are marked in green; down regulated genes are marked in red

Table 5 Topology table for up and down regulated genes

Construction of miRNA - target regulatory network

After combining the results of miRNA-target genes with the interactive network of miRNAs, 281 hub genes were selected and 2138 were miRNAs. The genes and miRNAs are shown in Fig. 4a. Specifically, 97 miRNAs (ex, hsa-mir-8067) that regulate RPL13A, 95 miRNAs (ex, hsa-mir-4518) that regulate RPS15A, 71 miRNAs (ex, hsa-mir-3685) that regulate RPLP0, 65 miRNAs (ex, hsa-mir-1202) that regulates ADCY6, 48 miRNAs (ex, hsa-mir-4461) that regulate RPS29, 129 miRNAs (ex, hsa-mir-8082) that regulate CTNND1, 98 miRNAs (ex, hsa-mir-4422) that regulate ESR1, 76 miRNAs (ex, hsa-mir-548am-5p) that regulate NEDD4L, 62 miRNAs (ex, hsa-mir-6886-3p) that regulate KNTC1 and 56 miRNAs (ex, hsa-mir-9500) that regulate NGFR were detected (Table 6).

Fig. 4
figure 4

a Target gene - miRNA regulatory network between target genes and miRNAs. b Target gene - TF regulatory network between target genes and TFs. Up regulated genes are marked in green; down regulated genes are marked in red; The purple color diamond nodes represent the key miRNAs; the blue color triangle nodes represent the key TFs.

Construction of TF - target regulatory network

After combining the results of TF-target genes with the interactive network of TFs, 455 hub genes were selected and 274 were TFs. The genes and TFs are shown in Fig. 4b. Specifically, 15 TFs (ex, PER3) that regulate RBX1, 13 TFs (ex, CTCF) that regulate RPS15, 12 TFs (ex, E2F7) that regulate RPS20, 11 TFs (ex, LMO2) that regulate ADCY6, 9 TFs (ex, POLR2H) that regulate POLR2K, 122 TFs (ex, NCOA2) that regulate ESR1, 21 miRNAs (ex, EBF1) that regulate LCK, 18 TFs (ex, SMAD2) that regulate GLI3, 17 TFs (ex, JUND) that regulate NEDD4L, and 15 TFs (ex, FOXO3) that regulate CALCR were detected (Table 6).

Receiver operating characteristic (ROC) curve analysis

Moreover, ROC curve analysis using “pROC” packages was performed to calculate the capacity of ten hub genes to distinguish PCOS from normal control. SAA1, ADCY6, POLR2K, RPS15, RPS15A, CTNND1, ESR1, NEDD4L, KNTC1 and NGFR all exhibited excellent diagnostic efficiency (AUC > 0.7) (Fig. 5).

Fig. 5
figure 5

ROC curve validated the sensitivity, specificity of hub genes as a predictive biomarker for PCOS prognosis. a SAA1, b ADCY6, c POLR2K, d RPS15, e RPS15A, f ESR1, g LCK, h S1PR5, i CCL28, j CTNND1

Validation of the expression levels of hub genes by RT-PCR

Aiming to further verify the expression patterns of selected hub genes, real-time PCR, which allows quantitative analysis of hub gene expression, was applied. The results showed that the relative expression levels of 10 hub genes including SAA1, ADCY6, POLR2K, RPS15, RPS15A, CTNND1, ESR1, NEDD4L, KNTC1 and NGFR were consistent with the expression profiling by high throughput sequencing (Fig. 6).

Fig. 6
figure 6

Validation of hub genes by RT- PCR. a SAA1, b ADCY6, c POLR2K, d RPS15, e RPS15A, f ESR1, g LCK, h S1PR5, i CCL28, j CTNND1

Molecular docking studies

In the present analysis, the docking simulations are performed to classify the active site conformation and significant interactions with the receptor binding sites responsible for complex stability. The over expressed genes is recognized in polycystic ovary syndrome and their x-ray crystallographic proteins structure are selected from PDB for docking studies. The standard drugs containing steroid nucleus are most commonly used either alone or in combination with other drugs. The docking studies of standard molecules containing the steroid ring have been carried out using Sybyl X 2.1 drug design software. The docking studies were performed to know the biding interaction of standard molecules on identified overexpressed genes of protein. The X- RAY crystallographic structure of one protein in each of four over expressed genes of POLR2K, RPS15, RPS15 and SAA1 of their co-crystallised protein of PDB code 1LE9, 3OW2, 1G1X and 4IP8 respectively were selected for the docking (Fig. 7). A total of three drug molecules of ethinylestradiol (ETE), levonorgestril (LNG) and desogestril (DSG) were docked with over expressed proteins to assess the binding affinity with proteins. The binding score greater than six are said to be good, all three drug molecules obtained binding score greater than 7 respectively. The molecules ETE obtained with a high binding score of 9.943 with SAA1 of PDB code 4IP8 and 8.260, 8.223 and 8.019 with 1G1X, 3OW2 and 1LE9. The LNG obtained highest binding score of 8.535 with SAA1 of PDB code 4IP8 and 8.351, 7.973 and 7.854 with RPS15, POLR2K and RPS15 alpha of PDB code 3OW2, 1LE9 and 1G1X respectively. DSG: highest with POLR2K of 8.273 with PDB code 1LE9, 8.158 with SAA1 of PDB code 4IP8, 7.745 with RPS15 alpha of PDB code 1G1X and obtained least binding score of 5.674 with RPS15 of PDB code 3OW2 respectively (Table 7). The molecule ETE and LNG has highest binding score its interaction with protein 4IP8 and hydrogen bonding and other bonding interactions with amino acids are depicted by 3D (Fig. 8) and 2D (Fig. 9)

Fig. 7
figure 7

Structures of Designed Molecules

Table 6 miRNA - target gene and TF - target gene interaction
Table 7 Docking results of standard drugs on overexpressed proteins
Fig. 8
figure 8

2D Binding of Molecule ETE with 4IP8

Fig. 9
figure 9

3D Binding of Molecule ETE with 4IP8

Discussion

PCOS is a most prevalent endocrine disorder with hyperandrogenism and chronic anovulation [29]. If not treated promptly and effectively, PCOS can seriously reduce the quality of life. There is no doubt that considerate syndrome at the molecular level will help to develop their diagnosis and treatment [30]. Up to now, various biomarkers have been identified to be linked with PCOS and might be selected as therapeutic targets, but the detailed mechanism of gene regulation leading to syndrome advancement remains elusive [31].

In our investigation, we aimed to identify biomarkers of PCOS and uncover their biological functions through bioinformatics analysis. Dataset GSE84958 was selected as expression profiling by high throughput sequencing dataset in our analysis. As a result, 360 up regulated and 379 down regulated genes at least 4-fold change between PCOS and normal control samples were screened out. ABI3BP protein expression in heart tissue was significantly related with cardiovascular disease [32], but this gene might be liable for progression of PCOS. Romo-Yáñez et al [33] have revealed the expression of BNIP3 was linked with diabetic in pregnancies, but this gene might be responsible for progression of PCOS. F13A1 is an essential regulatory factor to be associated in PCOS development [34]. An investigation has reported that the ITIH4 can promote non-alcoholic fatty liver disease [35], but this gene might be important for progression of PCOS. Da et al [36] have suggested that the TET3 is an important role in controlling type 2 diabetes progressions, but this gene might be key role in PCOS.

The GO and pathway enrichment analysis was of great importance for interpreting the molecular mechanisms of the key cellular activities in PCOS. RPS5 [37], RBM3 [38], BAK1 [39], NDUFC2 [40], NDUFS4 [41], NDUFS5 [42], UQCRFS1 [43], COX6B1 [44], NDUFA13 [45], PRMT1 [46], RDX (radixin) [47], EPHB4 [48], SYNE2 [49], DNAH5 [50], NEDD4L [51], PDE4B [52] and CTNND1 [53] plays a critical role in the process of cardiovascular disease, but these genes might be linked with development of PCOS. Ostergaard et al [54], Zi et al [55], Kunej et al [56], Van der Schueren et al [57], Jin et al [58], Emdad et al [59], Liu et al [60], Scherag et al [61], Shi and Long [62], Sharma et al [63], Parente et al [64], Saint-Laurent et al [65] and Lee [66] demonstrated that over expression of COA3, PHB (prohibitin), UQCRC1, COX4I1, IFI27, MTDH (metadherin), S100A16, SDCCAG8, GLI2, NTN1, NLGN2, FGFR3 and PTPRN2 could cause obesity, but these genes might be involved in progression of PCOS. Alsters et al [67], Lee et al [68], Shiffman et al [69], Yaghootkar et al [70], Rotroff et al [71], Cheng et al [72], Baig et al [73], Zhang et al [74], Lebailly et al [75], Ferris et al [76], Lempainen et al [77] and McCallum et al [78] presented that high expression of CPE (carboxypeptidase E), RPL13A, CERS2, CCND2, PRPF31, SARM1, PLD1, EPHA4, ARNTL2, BATF3, IKZF4 and MEN1 were associated with diabetes, but these genes might be linked with advancement of PCOS. Wang et al [79], Tian et al [80], Zhang et al [81] and Carr et al [82] demonstrated that over expression of ATP6AP2, FIS1, GRK4 and KCNQ4 were found to be substantially related to hypertension, but these genes might be essential for PCOS progression. Atiomo et al [83], Lara et al [84] and Douma et al [85] were reported that NQO1, NGFR (nerve growth factor receptor) and ESR1 could be an index for PCOS. Jin et al [86] presented that GLI3 was associated with non-alcoholic fatty liver disease, but this gene might be linked with development of PCOS.

In the present investigation, PPI network and its modules has been shown that significant amount of hub gene might be associated with progression of PCOS. Zhang et al [87] proposed that SAA1 was linked with progression of obesity, but this gene might be important for progression of PCOS. Deng et al [88] indicated that ADCY6 was responsible for development of cardiovascular disease, but this gene might be associated with advancement of PCOS. POLR2K, RPS15, RPS15A, ESR1, LCK (LCK proto-oncogene, Src family tyrosine kinase), S1PR5, CCL28, CTNND11, UQCRQ (ubiquinol-cytochrome c reductase complex III subunit VII), UQCRH (ubiquinol-cytochrome c reductase hinge protein), COX7C, COX6C, COX8A, COX5B, COX6A1, COX7A2L, ARHGAP39, OBSCN (obscurin, cytoskeletal calmodulin and titin-interacting RhoGEF) and TIAM2 might be novel biomarkers for PCOS.

MiRNA-target genes and TF-target genes regulatory networks revealed that the miRNAs, TF and target genes were might be involved in PCOS. Hsa-mir-6886-3p was liable for progression of hypertension [89], but this gene might be involved in progression of PCOS. Some investigations determined that expression of PER3 [90] and SMAD2 [91] were associated with diabetes, but these genes might be linked with advancement of PCOS. NCOA2 was found to be associated with advancement of obesity [92], but this gene might be involved in progression of PCOS. Recently, increasing evidence demonstrated that EBF1 was expressed in coronary artery disease [93], but this gene might be responsible for progression of PCOS. FOXO3 was involved in progression of PCOS [94]. RPLP0, RPS29, KNTC1, hsa-mir-8067, hsa-mir-4518, hsa-mir-3685, hsa-mir-1202, hsa-mir-4461, hsa-mir-8082, hsa-mir-4422, hsa-mir-548am-5p, hsa-mir-9500, RBX1, RPS20, CALCR (calcitonin receptor), CTCF (CCCTC-binding factor), E2F7, LMO2, POLR2H and JUND (jun D proto-oncogene) might be novel biomarkers for PCOS.

Among all three of molecules of ethinylestradiol, levonorgestrel and desogetril respectively, ethinylestradiolhas obtained highest binding score (c-score) of 9.943 with protein of PDB code 4IP8 and obtained 8.260, 8.223 and 8.019 with protein of PDB 1G1X, 3OW2 and 1LE9 respectively. The phenolic -OH group in ring A of ethinylestradiol formed favourable bonding interactions with ALA-14 of Chain A and pi-pi bonding interactions of alicyclic ring B TRP-18. Ethinylestradiol also formed alkyl and pi-alkyl interaction of ring B, C and D with TRP-18, ARG-62, TYR-21, PHE-69, ILE-65 and ILE-58. Ethinylestradiol also formed Van der Waals interactions with ACA-61, MET-17, MET-24 and GLN-66 respectively. It is assumed that the highest binding score (c-score) of ethinylestradiol is due to the presence of aromatic ring and the phenolic –OH group.

In conclusion, we used a series of bioinformatics analysis methods to find the crucial genes and pathways associated in PCOS initiation and development from expression profiling by high throughput sequencing containing PCOS samples and normal control samples. Our investigations provide a more specific molecular mechanism for the advancement of PCOS, detail information on the potential biomarkers and therapeutic targets. However, the interacting mechanism and function of genes need to be confirmed in further experiments.

Availability of data and materials

The datasets supporting the conclusions of this article are available in the GEO (http://www.ncbi.nlm.nih.gov/geo) repository. [(GSE84958) (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE84958)]

References

  1. Meier RK. Polycystic Ovary Syndrome. Nurs Clin North Am. 2018;53(3):407–20. https://doi.org/10.1016/j.cnur.2018.04.008.

    Article  PubMed  Google Scholar 

  2. Belenkaia LV, Lazareva LM, Walker W, Lizneva DV, Suturina LV. Criteria, phenotypes and prevalence of polycystic ovary syndrome. Minerva Ginecol 2019;71(3):211-223. doi:10.23736/S0026-4784.19.04404-6

  3. Escobar-Morreale HF, Roldán-Martín MB. Type 1 Diabetes and Polycystic Ovary Syndrome: Systematic Review and Meta-analysis. Diabetes Care. 2016;39(4):639–48. https://doi.org/10.2337/dc15-2577.

    Article  CAS  PubMed  Google Scholar 

  4. Oliver-Williams C, Vassard D, Pinborg A, Schmidt L. Risk of cardiovascular disease for women with polycystic ovary syndrome: results from a national Danish registry cohort study. Eur J Prev Cardiol. 2020:2047487320939674. https://doi.org/10.1177/2047487320939674.

  5. Lim SS, Norman RJ, Davies MJ, Moran LJ. The effect of obesity on polycystic ovary syndrome: a systematic review and meta-analysis. Obes Rev. 2013;14(2):95–109. https://doi.org/10.1111/j.1467-789X.2012.01053.x.

    Article  CAS  PubMed  Google Scholar 

  6. Wu J, Yao XY, Shi RX, Liu SF, Wang XY. A potential link between polycystic ovary syndrome and non-alcoholic fatty liver disease: an update meta-analysis. Reprod Health. 2018;15(1):77. https://doi.org/10.1186/s12978-018-0519-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Joham AE, Boyle JA, Zoungas S, Teede HJ. Hypertension in Reproductive-Aged Women With Polycystic Ovary Syndrome and Association With Obesity. Am J Hypertens. 2015;28(7):847–51. https://doi.org/10.1093/ajh/hpu251.

    Article  PubMed  Google Scholar 

  8. Forlenza GP, Calhoun A, Beckman KB, Halvorsen T, Hamdoun E, Zierhut H, Sarafoglou K, Polgreen LE, Miller BS, Nathan B, et al. Next generation sequencing in endocrine practice. Mol Genet Metab. 2015;115(2-3):61–71. https://doi.org/10.1016/j.ymgme.2015.05.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang LP, Peng XY, Lv XQ, Liu L, Li XL, He X, Lv F, Pan Y, Wang L, Liu KF, et al. High throughput circRNAs sequencing profile of follicle fluid exosomes of polycystic ovary syndrome patients. J Cell Physiol 2019. doi:10.1002/jcp.28201

  10. Clough E, Barrett T. The Gene Expression Omnibus Database. Methods Mol Biol. 2016;1418:93–110. https://doi.org/10.1007/978-1-4939-3578-9_5.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 2015;43(7):e47. doi:10.1093/nar/gkv007

  12. Ferreira JA. The Benjamini-Hochberg method in the case of discrete test statistics. Int J Biostat 2007;3(1):. doi:10.2202/1557-4679.1065

  13. Thomas PD. The Gene Ontology and the Meaning of Biological Function. Methods Mol Biol. 2017;1446:15–24. https://doi.org/10.1007/978-1-4939-3743-1_2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res 2009;37(Web Server issue):W305-W311. doi:10.1093/nar/gkp427

  15. Fabregat A, Jupe S, Matthews L, Sidiropoulos K, Gillespie M, Garapati P, Haw R, Jassal B, Korninger F, May B, et al. The Reactome Pathway Knowledgebase. Nucleic Acids Res. 2018;46(D1):D649–55. https://doi.org/10.1093/nar/gkx1132.

    Article  CAS  PubMed  Google Scholar 

  16. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43(Database issue):D447–52. https://doi.org/10.1093/nar/gku1003.

    Article  CAS  PubMed  Google Scholar 

  17. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B. Ideker T Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. https://doi.org/10.1101/gr.1239303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Przulj N, Wigle DA, Jurisica I. Functional topology in a network of protein interactions. Bioinformatics. 2004;20(3):340–8. https://doi.org/10.1093/bioinformatics/btg415.

    Article  CAS  PubMed  Google Scholar 

  19. Nguyen TP, Liu WC, Jordán F. Inferring pleiotropy by network analysis: linked diseases in the human PPI network. BMC Syst Biol 2011;5:179. Published 2011 Oct 31. doi:10.1186/1752-0509-5-179

  20. Shi Z, Zhang B. Fast network centrality analysis using GPUs. BMC Bioinformatics. 2011;12:149. https://doi.org/10.1186/1471-2105-12-149.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Fadhal E, Gamieldien J, Mwambene EC. Protein interaction networks as metric spaces: a novel perspective on distribution of hubs. BMC Syst Biol. 2014;8:6. https://doi.org/10.1186/1752-0509-8-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zaki N, Efimov D, Berengueres J. Protein complex detection using interaction reliability assessment and weighted clustering coefficient. BMC Bioinformatics. 2013;14:163. https://doi.org/10.1186/1471-2105-14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fan Y, Xia J. miRNet-Functional Analysis and Visual Exploration of miRNA-Target Interactions in a Network Context. Methods Mol Biol. 2018;1819:215–33. https://doi.org/10.1007/978-1-4939-8618-7_10.

    Article  CAS  PubMed  Google Scholar 

  24. Robin X, Turck N, Hainard A, Tiberti N, Lisacek F, Sanchez JC, Müller M. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics. 2011;12:77. https://doi.org/10.1186/1471-2105-12-77.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8. https://doi.org/10.1006/meth.2001.1262.

    Article  CAS  PubMed  Google Scholar 

  26. Shah KN, Patel SS. Phosphatidylinositide 3-kinase inhibition: A new potential target for the treatment of polycystic ovarian syndrome. Pharm Biol. 2016;54(6):975–83. https://doi.org/10.3109/13880209.2015.1091482.

    Article  CAS  PubMed  Google Scholar 

  27. Wang Y, Fu X, Xu J, Wang Q, Kuang H. Systems pharmacology to investigate the interaction of berberine and other drugs in treating polycystic ovary syndrome. Sci Rep 2016;6:28089. Published 2016 Jun 16. doi:10.1038/srep28089

  28. Wu XK, Zhou SY, Liu JX, Pöllänen P, Sallinen K, Mäkinen M, Erkkola R. Selective ovary resistance to insulin signaling in women with polycystic ovary syndrome. Fertil Steril. 2003;80(4):954–65. https://doi.org/10.1016/s0015-0282(03)01007-0.

    Article  PubMed  Google Scholar 

  29. Polak K, Czyzyk A, Simoncini T, Meczekalski B. New markers of insulin resistance in polycystic ovary syndrome. J Endocrinol Invest. 2017;40(1):1–8. https://doi.org/10.1007/s40618-016-0523-8.

    Article  CAS  PubMed  Google Scholar 

  30. Escobar-Morreale HF. Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nat Rev Endocrinol. 2018;14(5):270–84. https://doi.org/10.1038/nrendo.2018.24.

    Article  PubMed  Google Scholar 

  31. Carvalho LML, Ferreira CN, Sóter MO, Sales MF, Rodrigues KF, Martins SR, Candido AL, Reis FM, Silva IFO, Campos FMF, et al. Microparticles: Inflammatory and haemostatic biomarkers in Polycystic Ovary Syndrome. Mol Cell Endocrinol. 2017;443:155–62. https://doi.org/10.1016/j.mce.2017.01.017.

    Article  CAS  PubMed  Google Scholar 

  32. Delfín DA, DeAguero JL, McKown EN. The Extracellular Matrix Protein ABI3BP in Cardiovascular Health and Disease. Front Cardiovasc Med. 2019;6:23. https://doi.org/10.3389/fcvm.2019.00023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Romo-Yáñez J, Domínguez-Castro M, Flores-Reyes JS, Estrada-Juárez H, Mancilla-Herrera I, Hernández-Pineda J, Bazan-Tejeda ML, Aguinaga-Ríos M, Reyes-Muñoz E. Hyperglycemia differentially affects proliferation, apoptosis, and BNIP3 and p53 mRNA expression of human umbilical cord Wharton's jelly cells from non-diabetic and diabetic pregnancies. Biochem Biophys Res Commun. 2019;508(4):1149–54. https://doi.org/10.1016/j.bbrc.2018.12.037.

    Article  CAS  PubMed  Google Scholar 

  34. Schweighofer N, Lerchbaum E, Trummer O, Schwetz V, Pilz S, Pieber TR, Obermayer-Pietsch B. Androgen levels and metabolic parameters are associated with a genetic variant of F13A1 in women with polycystic ovary syndrome. Gene. 2012;504(1):133–9. https://doi.org/10.1016/j.gene.2012.04.050.

    Article  CAS  PubMed  Google Scholar 

  35. Nakamura N, Hatano E, Iguchi K, Sato M, Kawaguchi H, Ohtsu I, Sakurai T, Aizawa N, Iijima H, Nishiguchi S, et al. Elevated levels of circulating ITIH4 are associated with hepatocellular carcinoma with nonalcoholic fatty liver disease: from pig model to human study. BMC Cancer. 2019;19(1):621. https://doi.org/10.1186/s12885-019-5825-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Da Li, Cao T, Sun X, Jin S, Di Xie, Huang X, Yang X, Carmichael GG, Taylor HS, Diano S, et al. Hepatic TET3 contributes to type-2 diabetes by inducing the HNF4α fetal isoform. Nat Commun 2020;11(1):342. doi:10.1038/s41467-019-14185-z

  37. Zhang X, Hu C, Zhang N, Wei WY, Li LL, Wu HM, Ma ZG, Tang QZ. Matrine attenuates pathological cardiac fibrosis via RPS5/p38 in mice. Acta Pharmacol Sin 2020. doi:10.1038/s41401-020-0473-8

  38. Rosenthal LM, Leithner C, Tong G, Streitberger KJ, Krech J, Storm C, Schmitt KRL. RBM3 and CIRP expressions in targeted temperature management treated cardiac arrest patients-A prospective single center study. PLoS One. 2019;14(12):e0226005. https://doi.org/10.1371/journal.pone.0226005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hatchwell E. BAK1 gene variation and abdominal aortic aneurysms-variants are likely due to sequencing of a processed gene on chromosome 20. Hum Mutat. 2010;31(1):108–11. https://doi.org/10.1002/humu.21147.

    Article  CAS  PubMed  Google Scholar 

  40. Raffa S, Chin XLD, Stanzione R, Forte M, Bianchi F, Cotugno M, Marchitti S, Micaloni A, Gallo G, Schirone L, et al. The reduction of NDUFC2 expression is associated with mitochondrial impairment in circulating mononuclear cells of patients with acute coronary syndrome. Int J Cardiol. 2019;286:127–33. https://doi.org/10.1016/j.ijcard.2019.02.027.

    Article  PubMed  Google Scholar 

  41. Zhang H, Gong G, Wang P, Zhang Z, Kolwicz SC, Rabinovitch PS, Tian R, Wang W. Heart specific knockout of Ndufs4 ameliorates ischemia reperfusion injury. J Mol Cell Cardiol. 2018;123:38–45. https://doi.org/10.1016/j.yjmcc.2018.08.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Iwahana T, Okada S, Kanda M, Oshima M, Iwama A, Matsumiya G, Kobayashi Y. Novel myocardial markers GADD45G and NDUFS5 identified by RNA-sequencing predicts left ventricular reverse remodeling in advanced non-ischemic heart failure: a retrospective cohort study. BMC Cardiovasc Disord. 2020;20(1):116. https://doi.org/10.1186/s12872-020-01396-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gusic M, Schottmann G, Feichtinger RG, Du C, Scholz C, Wagner M, Mayr JA, Lee CY, Yépez VA, Lorenz N, et al. Bi-Allelic UQCRFS1 Variants Are Associated with Mitochondrial Complex III Deficiency, Cardiomyopathy, and Alopecia Totalis. Am J Hum Genet. 2020;106(1):102–11. https://doi.org/10.1016/j.ajhg.2019.12.005.

    Article  CAS  PubMed  Google Scholar 

  44. Abdulhag UN, Soiferman D, Schueler-Furman O, Miller C, Shaag A, Elpeleg O, Edvardson S, Saada A. Mitochondrial complex IV deficiency, caused by mutated COX6B1, is associated with encephalomyopathy, hydrocephalus and cardiomyopathy. Eur J Hum Genet. 2015;23(2):159–64. https://doi.org/10.1038/ejhg.2014.85.

    Article  CAS  PubMed  Google Scholar 

  45. Hu H, Nan J, Sun Y, Zhu D, Xiao C, Wang Y, Zhu L, Wu Y, Zhao J, Wu R, et al. Electron leak from NDUFA13 within mitochondrial complex I attenuates ischemia-reperfusion injury via dimerized STAT3. Proc Natl Acad Sci U S A. 2017;114(45):11908–13. https://doi.org/10.1073/pnas.1704723114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pyun JH, Kim HJ, Jeong MH, Ahn BY, Vuong TA, Lee DI, Choi S, Koo SH, Cho H, Kang JS. Cardiac specific PRMT1 ablation causes heart failure through CaMKII dysregulation. Nat Commun. 2018;9(1):5107. https://doi.org/10.1038/s41467-018-07606-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cetinkaya A, Berge B, Sen-Hild B, Troidl K, Gajawada P, Kubin N, Valeske K, Schranz D, Akintürk H, Schönburg M, et al. Radixin Relocalization and Nonmuscle α-Actinin Expression Are Features of Remodeling Cardiomyocytes in Adult Patients with Dilated Cardiomyopathy. Dis Markers. 2020;2020:9356738. https://doi.org/10.1155/2020/9356738.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yang D, Jin C, Ma H, Huang M, Shi GP, Wang J, Xiang M. EphrinB2/EphB4 pathway in postnatal angiogenesis: a potential therapeutic target for ischemic cardiovascular disease. Angiogenesis. 2016;19(3):297–309. https://doi.org/10.1007/s10456-016-9514-9.

    Article  CAS  PubMed  Google Scholar 

  49. Chen S, Wang C, Wang X, Xu C, Wu M, Wang P, Tu X, Wang QK. Significant Association Between CAV1 Variant rs3807989 on 7p31 and Atrial Fibrillation in a Chinese Han Population. J Am Heart Assoc. 2015;4(5):e001980. https://doi.org/10.1161/JAHA.115.001980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Nöthe-Menchen T, Wallmeier J, Pennekamp P, Höben IM, Olbrich H, Loges NT, Raidt J, Dougherty GW, Hjeij R, Dworniczak B, et al. Randomization of left-right Asymmetry and Congenital Heart defects: the role of DNAH5 in humans and mice. Circ Genom Precis Med 2019;doi:10.1161/CIRCGEN.119.002686

  51. Dahlberg J, Sjögren M, Hedblad B, Engström G, Melander O. Genetic variation in NEDD4L, an epithelial sodium channel regulator, is associated with cardiovascular disease and cardiovascular death. J Hypertens. 2014;32(2):294–9. https://doi.org/10.1097/HJH.0000000000000044.

    Article  CAS  PubMed  Google Scholar 

  52. Karam S, Margaria JP, Bourcier A, Mika D, Varin A, Bedioune I, Lindner M, Bouadjel K, Dessillons M, Gaudin F, et al. Cardiac Overexpression of PDE4B Blunts β-Adrenergic Response and Maladaptive Remodeling in Heart Failure. Circulation. 2020;142(2):161–74. https://doi.org/10.1161/CIRCULATIONAHA.119.042573.

    Article  CAS  PubMed  Google Scholar 

  53. Alharatani R, Ververi A, Beleza-Meireles A, Ji W, Mis E, Patterson QT, Griffin JN, Bhujel N, Chang CA, Dixit A, et al. Novel truncating mutations in CTNND1 cause a dominant craniofacial and cardiac syndrome. Hum Mol Genet. 2020;29(11):1900–21. https://doi.org/10.1093/hmg/ddaa050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ostergaard E, Weraarpachai W, Ravn K, Born AP, Jønson L, Duno M, Wibrand F, Shoubridge EA, Vissing J. Mutations in COA3 cause isolated complex IV deficiency associated with neuropathy, exercise intolerance, obesity, and short stature. J Med Genet. 2015;52(3):203–7. https://doi.org/10.1136/jmedgenet-2014-102914.

    Article  CAS  PubMed  Google Scholar 

  55. Zi Xu YX, Ande SR, Mishra S. Prohibitin: A new player in immunometabolism and in linking obesity and inflammation with cancer. Cancer Lett. 2018;415:208–16. https://doi.org/10.1016/j.canlet.2017.12.001.

    Article  CAS  PubMed  Google Scholar 

  56. Kunej T, Wang Z, Michal JJ, Daniels TF, Magnuson NS, Jiang Z. Functional UQCRC1 polymorphisms affect promoter activity and body lipid accumulation. Obesity. 2007;15(12):2896–901. https://doi.org/10.1038/oby.2007.344.

    Article  CAS  PubMed  Google Scholar 

  57. Van der Schueren B, Vangoitsenhoven R, Geeraert B, De Keyzer D, Hulsmans M, Lannoo M, Huber HJ, Mathieu C, Holvoet P. Low cytochrome oxidase 4I1 links mitochondrial dysfunction to obesity and type 2 diabetes in humans and mice. Int J Obes. 2015;39(8):1254–63. https://doi.org/10.1038/ijo.2015.58.

    Article  CAS  Google Scholar 

  58. Jin W, Jin W, Pan D. Ifi27 is indispensable for mitochondrial function and browning in adipocytes. Biochem Biophys Res Commun. 2018;501(1):273–9. https://doi.org/10.1016/j.bbrc.2018.04.234.

    Article  CAS  PubMed  Google Scholar 

  59. Emdad L, Das SK, Hu B, Kegelman T, Kang DC, Lee SG, Sarkar D, Fisher PB. AEG-1/MTDH/LYRIC: A Promiscuous Protein Partner Critical in Cancer, Obesity, and CNS Diseases. Adv Cancer Res. 2016;131:97–132. https://doi.org/10.1016/bs.acr.2016.05.002.

    Article  CAS  PubMed  Google Scholar 

  60. Liu Y, Zhang R, Xin J, Sun Y, Li J, Wei D, Zhao AZ. Identification of S100A16 as a novel adipogenesis promoting factor in 3T3-L1 cells. Endocrinology. 2011;152(3):903–11. https://doi.org/10.1210/en.2010-1059.

    Article  CAS  PubMed  Google Scholar 

  61. Scherag A, Kleber M, Boes T, Kolbe AL, Ruth A, Grallert H, Illig T, Heid IM, Toschke AM, Grau K, et al. SDCCAG8 obesity alleles and reduced weight loss after a lifestyle intervention in overweight children and adolescents. Obesity. 2012;20(2):466–70. https://doi.org/10.1038/oby.2011.339.

    Article  CAS  PubMed  Google Scholar 

  62. Shi Y, Long F. Hedgehog signaling via Gli2 prevents obesity induced by high-fat diet in adult mice. Elife. 2017;6:e31649. https://doi.org/10.7554/eLife.31649.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Sharma M, Schlegel M, Brown EJ, Sansbury BE, Weinstock A, Afonso MS, Corr EM, van Solingen C, Shanley LC, Peled D, et al. Netrin-1 Alters Adipose Tissue Macrophage Fate and Function in Obesity. Immunometabolism 2019;1(2):e190010. doi:10.20900/immunometab20190010

  64. Parente DJ, Garriga C, Baskin B, Douglas G, Cho MT, Araujo GC, Shinawi M. Neuroligin 2 nonsense variant associated with anxiety, autism, intellectual disability, hyperphagia, and obesity. Am J Med Genet A. 2017;173(1):213–6. https://doi.org/10.1002/ajmg.a.37977.

    Article  CAS  PubMed  Google Scholar 

  65. Saint-Laurent C, Garcia S, Sarrazy V, Dumas K, Authier F, Sore S, Tran A, Gual P, Gennero I, Salles JP, et al. Early postnatal soluble FGFR3 therapy prevents the atypical development of obesity in achondroplasia. PLoS One. 2018;13(4):e0195876. https://doi.org/10.1371/journal.pone.0195876.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Lee S. The association of genetically controlled CpG methylation (cg158269415) of protein tyrosine phosphatase, receptor type N2 (PTPRN2) with childhood obesity. Sci Rep. 2019;9(1):4855. https://doi.org/10.1038/s41598-019-40486-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Alsters SI, Goldstone AP, Buxton JL, Zekavati A, Sosinsky A, Yiorkas AM, Holder S, Klaber RE, Bridges N, van Haelst MM, et al. Truncating Homozygous Mutation of Carboxypeptidase E (CPE) in a Morbidly Obese Female with Type 2 Diabetes Mellitus, Intellectual Disability and Hypogonadotrophic Hypogonadism. PLoS One. 2015;10(6):e0131417. https://doi.org/10.1371/journal.pone.0131417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lee J, Harris AN, Holley CL, Mahadevan J, Pyles KD, Lavagnino Z, Scherrer DE, Fujiwara H, Sidhu R, Zhang J, et al. Rpl13a small nucleolar RNAs regulate systemic glucose metabolism. J Clin Invest. 2016;126(12):4616–25. https://doi.org/10.1172/JCI88069.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Shiffman D, Pare G, Oberbauer R, Louie JZ, Rowland CM, Devlin JJ, Mann JF, McQueen MJ. A gene variant in CERS2 is associated with rate of increase in albuminuria in patients with diabetes from ONTARGET and TRANSCEND. PLoS One. 2014;9(9):e106631. https://doi.org/10.1371/journal.pone.0106631.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yaghootkar H, Stancáková A, Freathy RM, Vangipurapu J, Weedon MN, Xie W, Wood AR, Ferrannini E, Mari A, Ring SM, et al. Association analysis of 29,956 individuals confirms that a low-frequency variant at CCND2 halves the risk of type 2 diabetes by enhancing insulin secretion. Diabetes. 2015;64(6):2279–85. https://doi.org/10.2337/db14-1456.

    Article  CAS  PubMed  Google Scholar 

  71. Rotroff DM, Yee SW, Zhou K, Marvel SW, Shah HS, Jack JR, Havener TM, Hedderson MM, Kubo M, Herman MA, et al. Genetic Variants in CPA6 and PRPF31 Are Associated With Variation in Response to Metformin in Individuals With Type 2 Diabetes. Diabetes. 2018;67(7):1428–40. https://doi.org/10.2337/db17-1164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Cheng Y, Liu J, Luan Y, Liu Z, Lai H, Zhong W, Yang Y, Yu H, Feng N, Wang H, et al. Sarm1 Gene Deficiency Attenuates Diabetic Peripheral Neuropathy in Mice. Diabetes. 2019;68(11):2120–30. https://doi.org/10.2337/db18-1233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Baig MH, Kausar MA, Husain FM, Shakil S, Ahmad I, Yadav BS, Saeed M. Interfering PLD1-PED/PEA15 interaction using self-inhibitory peptides: An in silico study to discover novel therapeutic candidates against type 2 diabetes. Saudi J Biol Sci. 2019;26(1):160–4. https://doi.org/10.1016/j.sjbs.2018.08.020.

    Article  CAS  PubMed  Google Scholar 

  74. Zhang Z, Tremblay J, Raelson J, Sofer T, Du L, Fang Q, Argos M, Marois-Blanchet FC, Wang Y, Yan L, et al. EPHA4 regulates vascular smooth muscle cell contractility and is a sex-specific hypertension risk gene in individuals with type 2 diabetes. J Hypertens. 2019;37(4):775–89. https://doi.org/10.1097/HJH.0000000000001948.

    Article  CAS  PubMed  Google Scholar 

  75. Lebailly B, He C, Rogner UC. Linking the circadian rhythm gene Arntl2 to interleukin 21 expression in type 1 diabetes. Diabetes. 2014;63(6):2148–57. https://doi.org/10.2337/db13-1702.

    Article  CAS  PubMed  Google Scholar 

  76. Ferris ST, Carrero JA, Mohan JF, Calderon B, Murphy KM, Unanue ER. A minor subset of Batf3-dependent antigen-presenting cells in islets of Langerhans is essential for the development of autoimmune diabetes. Immunity. 2014;41(4):657–69. https://doi.org/10.1016/j.immuni.2014.09.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Lempainen J, Härkönen T, Laine A, Knip M. Ilonen J; Finnish Pediatric Diabetes Register. Associations of polymorphisms in non-HLA loci with autoantibodies at the diagnosis of type 1 diabetes: INS and IKZF4 associate with insulin autoantibodies. Pediatr Diabetes. 2013;14(7):490–6. https://doi.org/10.1111/pedi.12046.

    Article  CAS  PubMed  Google Scholar 

  78. McCallum RW, Parameswaran V, Burgess JR. Multiple endocrine neoplasia type 1 (MEN 1) is associated with an increased prevalence of diabetes mellitus and impaired fasting glucose. Clin Endocrinol. 2006;65(2):163–8. https://doi.org/10.1111/j.1365-2265.2006.02563.x.

    Article  CAS  Google Scholar 

  79. Wang Y, Bao MH, Zhang QS, Li JM, Tang L. Association of ATP6AP2 Gene Polymorphisms with Essential Hypertension in a South Chinese Han Population. Asian Pac J Cancer Prev. 2015;16(17):8017–8. https://doi.org/10.7314/apjcp.2015.16.17.8017.

    Article  PubMed  Google Scholar 

  80. Tian L, Neuber-Hess M, Mewburn J, Dasgupta A, Dunham-Snary K, Wu D, Chen KH, Hong Z, Sharp WW, Kutty S, et al. Ischemia-induced Drp1 and Fis1-mediated mitochondrial fission and right ventricular dysfunction in pulmonary hypertension. J Mol Med. 2017;95(4):381–93. https://doi.org/10.1007/s00109-017-1522-8.

    Article  CAS  PubMed  Google Scholar 

  81. Zhang Y, Wang S, Huang H, Zeng A, Han Y, Zeng C, Zheng S, Ren H, Wang Y, Huang Y, et al. GRK4-mediated adiponectin receptor-1 phosphorylative desensitization as a novel mechanism of reduced renal sodium excretion in hypertension. Clin Sci. 2020;134(18):2453–67. https://doi.org/10.1042/CS20200671.

    Article  CAS  Google Scholar 

  82. Carr G, Barrese V, Stott JB, Povstyan OV, Jepps TA, Figueiredo HB, Zheng D, Jamshidi Y, Greenwood IA. MicroRNA-153 targeting of KCNQ4 contributes to vascular dysfunction in hypertension. Cardiovasc Res. 2016;112(2):581–9. https://doi.org/10.1093/cvr/cvw177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Atiomo W, Shafiee MN, Chapman C, Metzler VM, Abouzeid J, Latif A, Chadwick A, Kitson S, Sivalingam VN, Stratford IJ, et al. Corrigendum: Expression of NAD(P) H quinone dehydrogenase 1 (NQO1) is increased in the endometrium of women with endometrial cancer and women with polycystic ovary syndrome. Clin Endocrinol. 2017;87(6):886. https://doi.org/10.1111/cen.13515.

    Article  Google Scholar 

  84. Lara HE, Dissen GA, Leyton V, Paredes A, Fuenzalida H, Fiedler JL, Ojeda SR. An increased intraovarian synthesis of nerve growth factor and its low affinity receptor is a principal component of steroid-induced polycystic ovary in the rat. Endocrinology. 2000;141(3):1059–72. https://doi.org/10.1210/endo.141.3.7395.

    Article  CAS  PubMed  Google Scholar 

  85. Douma Z, Dallel M, Bahia W, Ben Salem A, Hachani B, Ali F, Almawi WY, Lautier C, Haydar S, Grigorescu F, Mahjoub T. Association of estrogen receptor gene variants (ESR1 and ESR2) with polycystic ovary syndrome in Tunisia. Gene 2020;741:144560. doi:10.1016/j.gene.2020.144560

  86. Jin SS, Lin XF, Zheng JZ, Wang Q, Guan HQ. lncRNA NEAT1 regulates fibrosis and inflammatory response induced by nonalcoholic fatty liver by regulating miR-506/GLI3. Eur Cytokine Netw. 2019;30(3):98–106. https://doi.org/10.1684/ecn.2019.0432.

    Article  CAS  PubMed  Google Scholar 

  87. Zhang X, Tang QZ, Wan AY, Zhang HJ, Wei L. SAA1 gene variants and childhood obesity in China. Lipids Health Dis. 2013;12:161. https://doi.org/10.1186/1476-511X-12-161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Deng Y, Wang J, Xie G, Zeng X, Li H. Circ-HIPK3 Strengthens the Effects of Adrenaline in Heart Failure by MiR-17-3p - ADCY6 Axis. Int J Biol Sci. 2019;15(11):2484–96. https://doi.org/10.7150/ijbs.36149.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Miao R, Wang Y, Wan J, Leng D, Gong J, Li J, Zhang Y, Pang W, Zhai Z, Yang Y, Miao R, Wang Y, Wan J, et al. Microarray Analysis and Detection of MicroRNAs Associated with Chronic Thromboembolic Pulmonary Hypertension. Biomed Res Int. 2017;2017:8529796. https://doi.org/10.1155/2017/8529796.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Karthikeyan R, Marimuthu G, Sooriyakumar M, BaHammam AS, Spence DW, Pandi-Perumal SR, Brown GM, Cardinali DP. Per3 length polymorphism in patients with type 2 diabetes mellitus. Horm Mol Biol Clin Investig. 2014;18(3):145–9. https://doi.org/10.1515/hmbci-2013-0049.

    Article  CAS  PubMed  Google Scholar 

  91. Lu Y, Habtetsion TG, Li Y, Zhang H, Qiao Y, Yu M, Tang Y, Zhen Q, Cheng Y, Liu Y. Association of NCOA2 gene polymorphisms with obesity and dyslipidemia in the Chinese Han population. Int J Clin Exp Pathol. 2015;8(6):7341–9.

    PubMed  PubMed Central  Google Scholar 

  92. Zhu Q, Chang A, Xu A, Luo K. The regulatory protein SnoN antagonizes activin/Smad2 protein signaling and thereby promotes adipocyte differentiation and obesity in mice. J Biol Chem. 2018;293(36):14100–11. https://doi.org/10.1074/jbc.RA118.003678.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Ying Y, Luo Y, Peng H. EBF1 gene polymorphism and its interaction with smoking and drinking on the risk of coronary artery disease for Chinese patients. Biosci Rep. 2018;38(3):BSR20180324. https://doi.org/10.1042/BSR20180324.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Zhang S, Deng W, Liu Q, Wang P, Yang W, Ni W. Altered m6 A modification is involved in up-regulated expression of FOXO3 in luteinized granulosa cells of non-obese polycystic ovary syndrome patients. J Cell Mol Med. 2020;24(20):11874–82. https://doi.org/10.1111/jcmm.15807.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgement

We thank Wiebke Arlt, University of Birmingham, Institute of Metabolism and Systems Research (IMSR), Birmingham, United Kingdom, very much, the author who deposited their microarray dataset, GSE84958, into the public GEO database.

Informed consent

No informed consent because this study does not contain human or animals participants.

Author information

Authors and Affiliations

Authors

Contributions

Praveenkumar Devarbhavi - Investigation and resources. Lata Telang - Writing original draft and investigation. Basavaraj Vastrad - Writing original draft, and review and editing. Anandkumar Tengli - Investigation, and review and editing. Chanabasayya Vastrad - Software and investigation. Iranna Kotturshetti - Supervision and resources. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Chanabasayya Vastrad.

Ethics declarations

Ethics approval and consent to participate

This article does not contain any studies with human participants or animals performed by any of the authors.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Devarbhavi, P., Telang, L., Vastrad, B. et al. Identification of key pathways and genes in polycystic ovary syndrome via integrated bioinformatics analysis and prediction of small therapeutic molecules. Reprod Biol Endocrinol 19, 31 (2021). https://doi.org/10.1186/s12958-021-00706-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12958-021-00706-3

Keywords