Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Identification of the shared gene signatures and pathways between polycystic ovary syndrome and endometrial cancer: An omics data based combined approach

  • Chenyun Miao ,

    Contributed equally to this work with: Chenyun Miao, Yun Chen, Xiaojie Fang

    Roles Data curation, Formal analysis, Writing – original draft

    Affiliation Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

  • Yun Chen ,

    Contributed equally to this work with: Chenyun Miao, Yun Chen, Xiaojie Fang

    Roles Project administration, Writing – review & editing

    Affiliation Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

  • Xiaojie Fang ,

    Contributed equally to this work with: Chenyun Miao, Yun Chen, Xiaojie Fang

    Roles Data curation, Software, Visualization

    Affiliation Department of Anorectal Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

  • Ying Zhao,

    Roles Supervision, Validation

    Affiliation Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

  • Ruye Wang,

    Roles Software

    Affiliation Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

  • Qin Zhang

    Roles Conceptualization, Methodology, Supervision

    zhaqin01@163.com

    Affiliation Department of TCM Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China

Abstract

Objective

Polycystic ovary syndrome (PCOS) is a common endocrine disorder with high incidence. Recently it has been implicated as a significant risk factor for endometrial cancer (EC). Our study aims to detect shared gene signatures and biological mechanism between PCOS and EC by bioinformatics analysis.

Methods

Bioinformatics analysis based on GEO database consisted of data integration, network construction and functional enrichment analysis was applied. In addition, the pharmacological methodology and molecular docking was also performed.

Results

Totally 10 hub common genes, MRPL16, MRPL22, MRPS11, RPL26L1, ESR1, JUN, UBE2I, MRPL17, RPL37A, GTF2H3, were considered as shared gene signatures for EC and PCOS. The GO and KEGG pathway analysis of these hub genes showed that “mitochondrial translational elongation”, “ribosomal subunit”, “structural constituent of ribosome” and “ribosome” were highly correlated. Besides, associated transcription factors (TFs) and miRNAs network were constructed. We identified candidate drug molecules including fenofibrate, cinnarizine, propanil, fenthion, clindamycin, chloramphenicol, demeclocycline, hydrochloride, azacitidine, chrysene and artenimol according to these hub genes. Molecular docking analysis verified a good binding interaction of fenofibrate against available targets (JUN, ESR1, UBE2I).

Conclusion

Gene signatures and regulatory biological pathways were identified through bioinformatics analysis. Moreover, the molecular mechanisms of these signatures were explored and potential drug molecules associated with PCOS and EC were screened out.

Introduction

Polycystic ovary syndrome (PCOS) is characterized by high incidence rate of 5–12% [1, 2] and is one of the most frequently occurring endocrine disorders in women of reproductive age. Characteristics of PCOS include oligo/anovulation, hyperandrogenism and polycystic ovaries, and is associated with heterogeneous clinical presentations such as menstrual irregularity, infertility, hirsutism and insulin resistance [3]. Aided by advances in research that help understand the biological processes implicated in PCOS, it also has been confirmed to have links to cancers in the endometrium, ovaries, kidneys, hematopoietic and pancreas system [4].

Endometrial cancer (EC) is the most common gynecologic cancer in the Western world with rising incidence and mortality [5]. It is estimated to lead to around 76000 deaths worldwide annually [6]. Published meta-analyses report that PCOS is a significant risk factor for EC [7], the results of which show that women with PCOS have 3-fold higher risk of developing EC compared with women without PCOS [7, 8]. Women aged less than 54 years have a significantly high risk for EC compared with elderly women (OR, 4.05) [9]. Features for PCOS such as obesity and anovulation can increase estrogen level and progesterone resistance, leading to development of endometrial hyperplasia and ultimately EC [10, 11]. Intricate relationship between EC and PCOS has been recognized for a number of years, but the exact pathomechanism mainly the genetic relationship between PCOS and EC remains unclear.

Gene expression profiles analysis and bioinformatic analysis using microarray data have been widely used to identify characteristic patterns of gene expression, dysregulated biological pathways, and gene interactome. In the current study, we utilized a range of bioinformatic approach to screen common genes and to explore transcriptional regulatory networks consist of transcription factors (TFs) and miRNAs between PCOS and EC to identify common molecular signatures and potential mechanisms. Finally, potential drug molecules were suggested. This study can help understand the molecular mechanism of this association and provide information for therapeutic strategy of PCOS patients with EC, which is of some clinical implications.

Material and methods

Retrieval of gene expression data

Microarray data were retrieved from the Gene Expression Omnibus (GEO) (http://www.ncbi.nih.gov/geo) [12]. GSE48301 dataset analyzed using Agilent GPL6244 platform [HuGene-1_0-st] comprised whole genome expression arrays of different endometrial cell populations obtained from PCOS women (n = 6) and healthy controls (n = 6). GSE115810 dataset analyzed using Agilent GPL96 platform [HG-U133A] comparing gene expression arrays from normal human endometrium (n = 3) with gene expression arrays from endometrial cancer of different grades (n = 24).

Identification of DEGs and shared gene signatures between EC and PCOS

GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r/) is a web-based analysis tool that uses GeoQuery and Limma R packages for data analysis [12]. Differentially expressed genes (DEGs) were analyzed using a p value < 0.05 as the cut-off criteria. Common DEGs between GSE48301 and GSE115810 datasets, which were potential genes associated with EC risk in women with PCOS were identified by R software (version 4.0.3) and visualized by the Venn diagram (http://bioinformatics.psb.ugent.be/webtools/Venn/).

Protein-protein interaction (PPI) network analysis and identification of hub targets

A PPI network was constructed using STRING tool (https://string-db.org/) to further explore the interaction between the overlapping DEGs [13]. All interaction evidence contributes to nodes in a given network is scored, resulting in an interaction score [14]. The minimum interaction score was set as greater than 0.4, and unconnected nodes in the network were removed. Further, key nodes within the PPI network were selected as hub genes using cytohubba plugin in Cytoscape software [15]. Hub genes were selected mainly based on their Maximal Clique Centrality (MCC) algorithm, which indicates essentiality of nodes in biological network [16]. Given a node v, the MCC of v is defined as MCC(v) = ∑C∈S(v)(|C|-1)!, where S(v) is the collection of maximal cliques which contain v, and (|C|-1)! is the product of all positive integers less than |C| [16].

Functional enrichment analysis

Gene Ontology (GO) [17] and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis [18] of hub genes were performed using ClusterProfiler package in R (version 4.0.3) to determine the biological functions and signaling pathways associated with the hub genes. GO enrichment analysis is comprised of three main categories included biological process (BP), cell component (CC) and molecular function (MF). A statistical threshold criterion at a p-value < 0.05 was chosen for selecting significantly enriched GO terms and pathways.

TFs-genes-miRNAs interaction network

Network analyst 3.0 tool (https://www.networkanalyst.ca/) is an online visual analytical platform for comprehensive gene expression profiling [19]. All hub genes were uploaded to network analyst to identify TFs and miRNAs that potentially regulated the hub genes. Genes-TFs network and genes-miRNAs network were also constructed using the cytohubba plugin in Cytoscape according to MCC score and degree.

Identification of drug candidates and molecular docking

DSigDB database comprises of 19531 genes and 17389 compounds and provides a direct link between genes and drugs for drug development studies and translational research [20]. DSigDB database is accessed through Enrichr (https://amp.pharm.mssm.edu/Enrichr/) webserver and is used for analysis of the relationship between drugs and potential targets. Hub genes were uploaded to the database to find potential drug molecules for PCOS and EC that target these genes. The compounds were then sorted based on the adjusted p value (p<0.05) and the combined score that calculated using the p-value and z-score computed by assessing the deviation from the expected rank [21].

To explore potential binding of the drug candidates to hub genes, the 3D structures of the drug molecules were obtained from PubChem. In addition, crystal structures of target proteins were retrieved from the RCSB protein data bank (http://www.rcsb.org/) [22]. Molecular docking was then performed using AutoDock Vina tools, and the results were visualized using PyMol 2.4.0 [23, 24].

Results

DEGs and common genes were identified between PCOS and EC

GSE48301 dataset was used to explore DEGs for PCOS. The findings showed that a total of 2437 DEGs were identified from GSE48301 dataset. In addition, 2391 DEGs associated with EC were identified from GSE115810 dataset. Identification of overlapping genes between PCOS and EC was performed using R software. Visualization using the Venn diagram showed 192 common genes in PCOS and EC (Fig 1).

thumbnail
Fig 1. Venn diagram of the intersections of PCOS and EC.

Intersections represent the differentially expressed genes in PCOS associated data series and EC associated data series.

https://doi.org/10.1371/journal.pone.0271380.g001

Identification of hub genes

The 192 common genes were submitted to STRING 11.0 database for construction of a medium confidence (score>0.4) PPI network. MCC score-based assessment was used to further identifythe hub genes using the cytohubba plugin. The top 10 genes including MRPL16, MRPL22, MRPS11, RPL26L1, ESR1, JUN, UBE2I, MRPL17, RPL37A, GTF2H3 were considered as hub genes (Table 1). The network comprised of 15 nodes and 30 edges (Fig 2).

thumbnail
Fig 2. Hub genes screening.

The color of the nodes are illustrated from red to yellow in descending order of MCC score. Gray lines highlight the interactions; line thickness refers to the interaction score provided by STRING.

https://doi.org/10.1371/journal.pone.0271380.g002

Functional enrichment analysis

The findings indicated that several GO terms were enriched by the hub genes including 124 BP terms, 31 CC terms and 30 MF terms. Analysis of individual modules showed that “mitochondrial translational elongation”, “ribosomal subunit”, and “structural constituent of ribosome” were the most significantly enriched terms (Fig 3). KEGG pathway analysis was performed to identify dysregulated pathways enriched by the hub genes identified for PCOS and EC. The findings for KEGG pathways analysis showed that only one pathway, ribosome, was significantly enriched (Fig 4).

thumbnail
Fig 3. GO enrichment analysis.

Shown are the top 10 most significantly enriched terms of each category based on p-value. The bubbles’ sizes are scaled according to the count of the potential targets enriched in the pathways. The bubbles are colored from red to blue in descending order of p-value. BP, biological process; CC, cell component; MF, molecular function.

https://doi.org/10.1371/journal.pone.0271380.g003

thumbnail
Fig 4. Selected KEGG pathways.

Red nodes represent hub genes of PCOS and EC.

https://doi.org/10.1371/journal.pone.0271380.g004

Transcriptional signatures

Genes-TFs and genes-miRNAs interaction networks were reconstructed using experimentally verified interactions in the NetworkAnalyst platform to explore transcriptional signatures and post-transcriptional regulatory signatures [19]. There were 298 nodes and 316 edges in the genes-miRNAs network and 171 nodes and 244 edges in the genes-TFs network. Four TFs showed strong correlation with the hub genes namely, KLF9, PHF8, KDM5B, and SAP30(Fig 5). Nevertheless, no significantly correlated miRNAs were screened out in the cytohubba, MCC scores of all the related miRNAs were in the range of 1–2 (Fig 6).

thumbnail
Fig 5. Genes-TFs interaction network.

Hexagons represent hub genes; circle nodes represent TFs associated with hub genes.

https://doi.org/10.1371/journal.pone.0271380.g005

thumbnail
Fig 6. Genes-miRNAs interaction network.

Hexagons represent hub genes; circle nodes represent miRNAs associated with hub genes.

https://doi.org/10.1371/journal.pone.0271380.g006

Candidate small drug molecules

The identified hub genes for PCOS and EC were uploaded to Enrichr platform. The platform provides a list of potential molecules that target the genes based on data from DSigDB database. The top ten candidate drug molecules were generated after manually removing duplicates based on the adjusted p-value. The drug molecules were fenofibrate, cinnarizine, propanil, fenthion, clindamycin, chloramphenicol, demeclocycline, hydrochloride, azacitidine, chrysene and artenimol (Table 2). Fenofibrate was with the highest combined score.

thumbnail
Table 2. Candidate drug molecules (top ten) identified from gene-drug interaction enrichment analysis.

https://doi.org/10.1371/journal.pone.0271380.t002

Molecular docking analysis

Molecular docking was performed to evaluate the binding affinity of fenofibrate to 10 hub targets. A lower affinity score indicates stronger binding ability. The crystal structures of MRPL16, MRPL17, RPL26L1, MRPL22, RPL37A and GTF2H3 were not available in PDB database, thus molecular docking analysis was only performed for JUN(PDB ID:5FV8), ESR1(PDB ID:3UUD) and UBE2I(PDB ID:5F6E). Docking affinity scores for fenofibrate against JUN, ESR1 and UBE2I were all less than -1.2 kcal/mol implying that these compounds have reasonable binding affinities with the hub proteins (Fig 7 and Table 3).

thumbnail
Fig 7. Molecular docking of fenofibrate with proteins of hub genes.

(A). The binding poses of ESR1 complexed with fenofibrate. (B). The binding poses of JUN complexed with fenofibrate. (C). The binding poses of UBE2I complexed with fenofibrate. Hydrogen bonds are indicated as dashed lines.

https://doi.org/10.1371/journal.pone.0271380.g007

Discussion

EC is the most common malignancy type in females in the developed world and is associated with high incidence and mortality rate [25]. Approximately 60,000 females are diagnosed with EC, and 10,000 deaths are recorded each year [26]. Accordingly, in order to prevent it, identifying women at high risk of EC is important. Women with PCOS presenting with a 9% lifetime risk of EC are considered as a high-risk group for EC. Several clinical features of PCOS including obesity, insulin resistance, unregulated estrogen stimulation of the endometrium, diabetes and progesterone resistance are metabolic and molecular risk factors for EC [8]. However, the exact relationship between PCOS and EC has not been fully elucidated.

In the present study, bioinformatics analyses were used to identify hub genes for PCOS and EC, and to explore the transcriptional regulatory signatures for these genes. Notably, a total of 10 hub genes namely, MRPL16, MRPL22, MRPS11, RPL26L1, ESR1, JUN, UBE2I, MRPL17, RPL37A and GTF2H3 were identified from the DEGs of PCOS and EC endometrial tissues. GO analysis and KEGG pathway analysis, construction of genes-TFs and genes-miRNAs interaction networks, and small molecule drug prediction were performed to further explore the role of the hub genes. The finding showed that ribosome and mitochondrial translation were the most important common pathways for PCOS and EC, and ten drug molecules led by fenofibrate was detected as potential drugs to decrease EC risk for PCOS patients (Figs 8 and 9).

thumbnail
Fig 9. Brief schematic diagram.

A speculative mechanism diagram of the hub genes in PCOS endometrium carcinogenesis.

https://doi.org/10.1371/journal.pone.0271380.g009

KEGG pathway enrichment analysis results showed that ribosome associated with MRPL16, MRPL22, MRPS11, RPL26L1 MRPL17, RPL37A plays a key role in pathogenesis of PCOS and EC. Ribosome is a complex organelle involved in mRNA translation and protein synthesis [27]. Abundant findings indicate that hyperactive ribosome biogenesis promotes tumorigenesis through quantitative and qualitative changes in ribosomes that affect the process of translation [28]. Additionally, GO terms related to hub genes reveal that mitochondrial translation is implicated in the cancerous process in endometrial tissue of PCOS. Translation of mRNA involved in transcription of mtDNA into protein in mitochondrial ribosome comprises initiation, elongation and termination steps, which are regulated by several mammalian mitochondrial translation factors such as mtIF2, mtIF3, and mtEF4 [29]. Translation process in mitoribosomes is linked to oxidative phosphorylation (OXPHOS) defects and results in increased oxidant stress [30, 31]. Recent studies report that alteration in the level of mitoribosome translation is implicated in development and progression of tumors [32]. Notably, components of the mitochondrial translational machinery are potential targets for tumor treatment. For instance, inhibition of the mitochondrial translation elongation factor, EF-Tu in acute myeloid leukemia attenuates cell growth and improves tigecycline sensitivity [33]. A previous study using 42 patient biopsies reported that MRPS18-2 was significantly highly expressed in EC compared with the level in normal endometrium [34]. However, no evidence has explored the relationship between mitoribosome translation and PCOS. Further studies are strongly recommended to explore the detailed mechanism of mitoribosome translation in PCOS and EC.

TFs control gene expression by directly binding to DNA sequences of target genes thus play a regulatory role in transcription and translation processes [35, 36]. Moreover, miRNAs modulate mRNA translation and transcript degradation [37]. TFs and miRNAs modulate genetic expressions which may result in formation of cancer cells [38]. The hub genes identified in the current study were uploaded in network analyst platform for analysis of TF-genes interaction networks to identify TF associated with PCOS and EC. Analysis of the network showed that KLF9, PHF8, KDM5B and SAP30 were significantly correlated TFs with hub genes. They have all been shown to take part in tumorigenesis [3942]. Moreover, previous studies have suggested that KLF9 is a tumor suppressor involved in development of EC [43].

DSigDB database was used to identify potential molecular drugs for the 10 hub genes. The findings showed that fenofibrate was potential drug molecule for the hub genes. Fenofibrate is PPARα agonist and is widely applied in clinical practice as an effective lipid-lowering agent. Fenofibrate exerts its activity by increasing HDL levels and decreasing the levels of LDL, cholesterol and triglycerides [44]. Hyperlipidemia is a common feature in women with PCOS and is associated with several clinical characteristics of PCOS such as IR, hyperandrogenemia, anovulation and inflammation. Although it is not the first-line treatment for lipid-lowering in PCOS, fenofibrate is recommended owing to few drug interactions and low muscle toxicity [45]. Fenofibrate was recently reported to exert anticancer effects in various of human tumors [4648]. A previous study reported that fenofibrate inhibited proliferation and induced apoptosis in Ishikawa endometrial cancer cells [49]. Moreover, it promotes metabolism of fatty acids over glucose for the metabolic needs in tumor microenvironment thus decreasing tumor progression [50]. In order to explore the therapeutic potential of fenofibrate, we started a preliminary validation through molecular docking, which can predict binding affinities between molecule and protein residues using binding free energy (ΔGbind) [51]. Findings from molecular docking showed that fenofibrate interacted directly with the active residues of the target proteins including JUN, ESR1 and UBE2I via multiple hydrogen bonds. These findings indicate that fenofibrate is a potential candidate for future drug development targeting both EC and PCOS. Future experimental studies are required to test its potential in the treatment of PCOS patients with EC.

Conclusion

Currently, the relationship between PCOS and EC has not been completely understood. This is the first study to explore the association between PCOS and EC using an omics data based combined approach. Common DEGs were identified by screening genome expression data of different endometrial cells. Gene signatures and regulatory signatures were identified through bioinformatics analysis. Moreover, the molecular mechanisms of these signatures were explored and potential small drug molecules associated with the hub genes were identified. Further experimental and clinical studies should be conducted to verify the identified molecular signatures and potential drugs.

Supporting information

S2 Table. GEO dataset of endometrial cancer patients.

https://doi.org/10.1371/journal.pone.0271380.s002

(XLS)

S4 Table. Transcription factors (TFs) that regulate hub genes.

https://doi.org/10.1371/journal.pone.0271380.s004

(XLS)

Acknowledgments

We would like to thank Home for Researchers editorial team for English language editing.

References

  1. 1. Varanasi LC, Subasinghe A, Jayasinghe YL, Callegari ET, Garland SM, Gorelik A, et al. Polycystic ovarian syndrome: Prevalence and impact on the wellbeing of Australian women aged 16–29 years. Aust N Z J Obstet Gynaecol. 2018;58(2):222–33. Epub 2017/10/21. pmid:29052216.
  2. 2. Azziz R, Woods KS, Reyna R, Key TJ, Knochenhauer ES, Yildiz BO. The prevalence and features of the polycystic ovary syndrome in an unselected population. J Clin Endocrinol Metab. 2004;89(6):2745–9. Epub 2004/06/08. pmid:15181052.
  3. 3. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic ovary syndrome (PCOS). Hum Reprod. 2004;19(1):41–7. Epub 2003/12/23. pmid:14688154.
  4. 4. Yin W, Falconer H, Yin L, Xu L, Ye W. Association Between Polycystic Ovary Syndrome and Cancer Risk. JAMA Oncol. 2019;5(1):106–7. Epub 2018/11/30. pmid:30489606; PubMed Central PMCID: PMC6439760.
  5. 5. Kitson SJ, Rosser M, Fischer DP, Marshall KM, Clarke RB, Crosbie EJ. Targeting Endometrial Cancer Stem Cell Activity with Metformin Is Inhibited by Patient-Derived Adipocyte-Secreted Factors. Cancers. 2019;11(5). Epub 2019/05/15. pmid:31083574; PubMed Central PMCID: PMC6562824.
  6. 6. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. Epub 2018/09/13. pmid:30207593.
  7. 7. Chittenden BG, Fullerton G, Maheshwari A, Bhattacharya S. Polycystic ovary syndrome and the risk of gynaecological cancer: a systematic review. Reprod Biomed Online. 2009;19(3):398–405. Epub 2009/09/26. pmid:19778486.
  8. 8. Haoula Z, Salman M, Atiomo W. Evaluating the association between endometrial cancer and polycystic ovary syndrome. Hum Reprod. 2012;27(5):1327–31. Epub 2012/03/01. pmid:22367984.
  9. 9. Barry JA, Azizia MM, Hardiman PJ. Risk of endometrial, ovarian and breast cancer in women with polycystic ovary syndrome: a systematic review and meta-analysis. Hum Reprod Update. 2014;20(5):748–58. Epub 2014/04/02. pmid:24688118; PubMed Central PMCID: PMC4326303.
  10. 10. Raglan O, Kalliala I, Markozannes G, Cividini S, Gunter MJ, Nautiyal J, et al. Risk factors for endometrial cancer: An umbrella review of the literature. Int J Cancer. 2019;145(7):1719–30. Epub 2018/11/06. pmid:30387875.
  11. 11. Huang X, Zhong R, He X, Deng Q, Peng X, Li J, et al. Investigations on the mechanism of progesterone in inhibiting endometrial cancer cell cycle and viability via regulation of long noncoding RNA NEAT1/microRNA-146b-5p mediated Wnt/β-catenin signaling. IUBMB Life. 2019;71(2):223–34. Epub 2018/11/20. pmid:30452118.
  12. 12. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets—update. Nucleic Acids Res. 2013;41(Database issue):D991–5. Epub 2012/11/30. pmid:23193258; PubMed Central PMCID: PMC3531084.
  13. 13. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–d13. Epub 2018/11/27. pmid:30476243; PubMed Central PMCID: PMC6323986.
  14. 14. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605–d12. Epub 2020/11/26. pmid:33237311; PubMed Central PMCID: PMC7779004.
  15. 15. Bai Q, Liu H, Guo H, Lin H, Song X, Jin Y, et al. Identification of Hub Genes Associated With Development and Microenvironment of Hepatocellular Carcinoma by Weighted Gene Co-expression Network Analysis and Differential Gene Expression Analysis. Front Genet. 2020;11:615308. Epub 2021/01/09. pmid:33414813; PubMed Central PMCID: PMC7783465.
  16. 16. Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC systems biology. 2014;8 Suppl 4(Suppl 4):S11. Epub 2014/12/19. pmid:25521941; PubMed Central PMCID: PMC4290687.
  17. 17. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 2019;47(D1):D330–d8. Epub 2018/11/06. pmid:30395331; PubMed Central PMCID: PMC6323945.
  18. 18. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30. Epub 1999/12/11. pmid:10592173; PubMed Central PMCID: PMC102409.
  19. 19. Zhou G, Soufan O, Ewald J, Hancock REW, Basu N, Xia J. NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res. 2019;47(W1):W234–w41. Epub 2019/04/02. pmid:30931480; PubMed Central PMCID: PMC6602507.
  20. 20. Yoo M, Shin J, Kim J, Ryall KA, Lee K, Lee S, et al. DSigDB: drug signatures database for gene set analysis. Bioinformatics. 2015;31(18):3069–71. Epub 2015/05/21. pmid:25990557; PubMed Central PMCID: PMC4668778.
  21. 21. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128. Epub 2013/04/17. pmid:23586463; PubMed Central PMCID: PMC3637064.
  22. 22. Goodsell DS, Zardecki C, Di Costanzo L, Duarte JM, Hudson BP, Persikova I, et al. RCSB Protein Data Bank: Enabling biomedical research and drug discovery. Protein Sci. 2020;29(1):52–65. Epub 2019/09/19. pmid:31531901; PubMed Central PMCID: PMC6933845.
  23. 23. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31(2):455–61. Epub 2009/06/06. pmid:19499576; PubMed Central PMCID: PMC3041641.
  24. 24. Seeliger D, de Groot BL. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J Comput Aided Mol Des. 2010;24(5):417–22. Epub 2010/04/20. pmid:20401516; PubMed Central PMCID: PMC2881210.
  25. 25. de Haydu C, Black JD, Schwab CL, English DP, Santin AD. An update on the current pharmacotherapy for endometrial cancer. Expert Opin Pharmacother. 2016;17(4):489–99. Epub 2015/12/03. pmid:26629895.
  26. 26. Njoku K, Abiola J, Russell J, Crosbie EJ. Endometrial cancer prevention in high-risk women. Best Pract Res Clin Obstet Gynaecol. 2020;65:66–78. Epub 2020/02/29. pmid:32107136.
  27. 27. Noller HF. Evolution of protein synthesis from an RNA world. Cold Spring Harbor perspectives in biology. 2012;4(4):a003681. Epub 2010/07/09. pmid:20610545; PubMed Central PMCID: PMC3312679.
  28. 28. Pelletier J, Thomas G, Volarević S. Ribosome biogenesis in cancer: new players and therapeutic avenues. Nature reviews Cancer. 2018;18(1):51–63. Epub 2017/12/02. pmid:29192214.
  29. 29. D’Souza AR, Minczuk M. Mitochondrial transcription and translation: overview. Essays in biochemistry. 2018;62(3):309–20. Epub 2018/07/22. pmid:30030363; PubMed Central PMCID: PMC6056719.
  30. 30. Kim HJ, Maiti P, Barrientos A. Mitochondrial ribosomes in cancer. Seminars in cancer biology. 2017;47:67–81. Epub 2017/04/27. pmid:28445780; PubMed Central PMCID: PMC5662495.
  31. 31. Ashton TM, McKenna WG, Kunz-Schughart LA, Higgins GS. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clinical cancer research: an official journal of the American Association for Cancer Research. 2018;24(11):2482–90. Epub 2018/02/09. pmid:29420223.
  32. 32. Koc EC, Haciosmanoglu E, Claudio PP, Wolf A, Califano L, Friscia M, et al. Impaired mitochondrial protein synthesis in head and neck squamous cell carcinoma. Mitochondrion. 2015;24:113–21. Epub 2015/08/05. pmid:26238294.
  33. 33. Skrtić M, Sriskanthadevan S, Jhas B, Gebbia M, Wang X, Wang Z, et al. Inhibition of mitochondrial translation as a therapeutic strategy for human acute myeloid leukemia. Cancer cell. 2011;20(5):674–88. Epub 2011/11/19. pmid:22094260; PubMed Central PMCID: PMC3221282.
  34. 34. Mints M, Mushtaq M, Iurchenko N, Kovalevska L, Stip MC, Budnikova D, et al. Mitochondrial ribosomal protein S18-2 is highly expressed in endometrial cancers along with free E2F1. Oncotarget. 2016;7(16):22150–8. Epub 2016/03/10. pmid:26959119; PubMed Central PMCID: PMC5008351.
  35. 35. Le TD, Liu L, Zhang J, Liu B, Li J. From miRNA regulation to miRNA-TF co-regulation: computational approaches and challenges. Brief Bioinform. 2015;16(3):475–96. Epub 2014/07/14. pmid:25016381.
  36. 36. Mohanta TK, Yadav D, Khan A, Hashem A, Tabassum B, Khan AL, et al. Genomics, molecular and evolutionary perspective of NAC transcription factors. PLoS One. 2020;15(4):e0231425. Epub 2020/04/11. pmid:32275733; PubMed Central PMCID: PMC7147800.
  37. 37. Choudhari JK, Verma MK, Choubey J, Sahariah BP. Investigation of MicroRNA and transcription factor mediated regulatory network for silicosis using systems biology approach. Sci Rep. 2021;11(1):1265. Epub 2021/01/16. pmid:33446673; PubMed Central PMCID: PMC7809153.
  38. 38. Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nature medicine. 2004;10(8):789–99. Epub 2004/08/03. pmid:15286780.
  39. 39. Tseng L, Cheng H, Yeh T, Huang S, Syu Y, Chuu C, et al. Targeting the histone demethylase PHF8-mediated PKCα-Src-PTEN axis in HER2-negative gastric cancer. Proceedings of the National Academy of Sciences of the United States of America. 2020;117(40):24859–66. pmid:32958674.
  40. 40. Sun J, Wang B, Liu Y, Zhang L, Ma A, Yang Z, et al. Transcription factor KLF9 suppresses the growth of hepatocellular carcinoma cells in vivo and positively regulates p53 expression. Cancer letters. 2014;355(1):25–33. pmid:25242357.
  41. 41. Li G, Kanagasabai T, Lu W, Zou M, Zhang S, Celada S, et al. KDM5B Is Essential for the Hyperactivation of PI3K/AKT Signaling in Prostate Tumorigenesis. Cancer research. 2020;80(21):4633–43. pmid:32868382.
  42. 42. Sironi E, Cerri A, Tomasini D, Sirchia S, Porta G, Rossella F, et al. Loss of heterozygosity on chromosome 4q32-35 in sporadic basal cell carcinomas: evidence for the involvement of p33ING2/ING1L and SAP30 genes. Journal of cutaneous pathology. 2004;31(4):318–22. pmid:15005689.
  43. 43. Yin X, Li X, Feng G, Qu Y, Wang H. LINC00565 Enhances Proliferative Ability in Endometrial Carcinoma by Downregulating KLF9. Onco Targets Ther. 2020;13:6181–9. Epub 2020/07/09. pmid:32636642; PubMed Central PMCID: PMC7334012.
  44. 44. Lian X, Wang G, Zhou H, Zheng Z, Fu Y, Cai L. Anticancer Properties of Fenofibrate: A Repurposing Use. J Cancer. 2018;9(9):1527–37. Epub 2018/05/16. pmid:29760790; PubMed Central PMCID: PMC5950581.
  45. 45. Wild RA, Carmina E, Diamanti-Kandarakis E, Dokras A, Escobar-Morreale HF, Futterweit W, et al. Assessment of cardiovascular risk and prevention of cardiovascular disease in women with the polycystic ovary syndrome: a consensus statement by the Androgen Excess and Polycystic Ovary Syndrome (AE-PCOS) Society. J Clin Endocrinol Metab. 2010;95(5):2038–49. Epub 2010/04/09. pmid:20375205.
  46. 46. Liu H, Zang C, Fenner MH, Liu D, Possinger K, Koeffler HP, et al. Growth inhibition and apoptosis in human Philadelphia chromosome-positive lymphoblastic leukemia cell lines by treatment with the dual PPARalpha/gamma ligand TZD18. Blood. 2006;107(9):3683–92. Epub 2006/01/13. pmid:16403907.
  47. 47. Muzio G, Maggiora M, Oraldi M, Trombetta A, Canuto RA. PPARalpha and PP2A are involved in the proapoptotic effect of conjugated linoleic acid on human hepatoma cell line SK-HEP-1. Int J Cancer. 2007;121(11):2395–401. Epub 2007/08/11. pmid:17691108.
  48. 48. Shigeto T, Yokoyama Y, Xin B, Mizunuma H. Peroxisome proliferator-activated receptor alpha and gamma ligands inhibit the growth of human ovarian cancer. Oncol Rep. 2007;18(4):833–40. Epub 2007/09/06. pmid:17786343.
  49. 49. Saidi SA, Holland CM, Charnock-Jones DS, Smith SK. In vitro and in vivo effects of the PPAR-alpha agonists fenofibrate and retinoic acid in endometrial cancer. Mol Cancer. 2006;5:13. Epub 2006/03/30. pmid:16569247; PubMed Central PMCID: PMC1475879.
  50. 50. Chekaoui A, Ertl HCJ. PPARα Agonist Fenofibrate Enhances Cancer Vaccine Efficacy. Cancer Res. 2021;81(17):4431–40. Epub 2021/07/11. pmid:34244236.
  51. 51. Pinzi L, Rastelli G. Molecular Docking: Shifting Paradigms in Drug Discovery. Int J Mol Sci. 2019;20(18). Epub 2019/09/07. pmid:31487867; PubMed Central PMCID: PMC6769923.