Skip to main content

Bioinformatics analysis to screen the key prognostic genes in ovarian cancer

Abstract

Background

Ovarian cancer (OC) is a gynecological oncology that has a poor prognosis and high mortality. This study is conducted to identify the key genes implicated in the prognosis of OC by bioinformatic analysis.

Methods

Gene expression data (including 568 primary OC tissues, 17 recurrent OC tissues, and 8 adjacent normal tissues) and the relevant clinical information of OC patients were downloaded from The Cancer Genome Atlas database. After data preprocessing, cluster analysis was conducted using the ConsensusClusterPlus package in R. Using the limma package in R, differential analysis was performed to identify feature genes. Based on Kaplan-Meier (KM) survival analysis, prognostic seed genes were selected from the feature genes. After key prognostic genes were further screened by cluster analysis and KM survival analysis, they were performed functional enrichment analysis and multivariate survival analysis. Using the survival package in R, cox regression analysis was conducted for the microarray data of GSE17260 to validate the key prognostic genes.

Results

A total of 3668 feature genes were obtained, among which 75 genes were identified as prognostic seed genes. Then, 25 key prognostic genes were screened, including AXL, FOS, KLF6, WDR77, DUSP1, GADD45B, and SLIT3. Especially, AXL and SLIT3 were enriched in ovulation cycle. Multivariate survival analysis showed that the key prognostic genes could effectively differentiate the samples and were significantly associated with prognosis. Additionally, GSE17260 confirmed that the key prognostic genes were associated with the prognosis of OC.

Conclusion

AXL, FOS, KLF6, WDR77, DUSP1, GADD45B, and SLIT3 might affect the prognosis of OC.

Background

Ovarian cancer (OC), which ranks seventh in incidence and eighth in mortality among tumors in women, is characterized by pelvic pain, bloating, loss of appetite, and abdominal swelling [1]. OC can mainly spread into the lining of bowel and abdomen, and lymph nodes, bladder, liver, and lungs [2]. Usually, the women with more ovulation have higher risk of OC, especially those who have not given birth, have earlier menstruation or later menopause [3]. OC usually has a poor prognosis and high mortality, and most cases are diagnosed at advanced stages as there lacks effective detection means [4]. In 2012, globally 239,000 women were diagnosed with OC and nearly around 152,000 women died of the disease. [3]. Thus, exploring the pathogenesis of OC and developing novel therapies are urgent.

In recent years, several studies have reported the molecular mechanisms of OC. For instance, Li et al. declared that chemokine receptor 4 (CXCR4) plays a critical role in cisplatin-based chemotherapy for patients with epithelial ovarian cancer (EOC) and can be seen as a prognostic factor [5]. Yes-associated protein 1 (YAP) contributes to cell growth and formation of OC both in vivo and in vitro, additionally, YAP and TEA domain family member 4 (TEAD4) may serve as prognostic markers and therapeutic targets for OC [6, 7]. Previous studies demonstrated that high Beclin 1 expression in protein level can be a prognostic factor of OC [8, 9]. Califano et al. deemed that obesity evaluated by Body Mass Index combined with high mobility group A2 (HMGA2) expression can be used for predicting poor prognosis in patients suffered from OC [10]. Forkhead box M1 (FOXM1) expression is reported to be participated in the development and progression of EOC, and FOXM1 is a promising prognostic factor for overall survival and progression-free survival [11, 12]. However, there lacks a overall reveal of the key genes implicated in OC.

To identify the key genes associated with prognosis of OC, microarray data of primary OC tissues, recurrent OC tissues and adjacent normal tissues were obtained. Then, the samples were pre-classified into two groups, and key prognostic genes were screened. Followed by functional enrichment analysis, multivariate survival analysis was carried out to examine the overall influence of these genes on prognosis. Finally, the key prognostic genes were validated by an independent microarray data.

Methods

Data source and data preprocessing

Gene expression data of OC patients (dataset ID: TCGA_OV_exp_u133a) were downloaded from TCGA (The Cancer Genome Atlas, http://cancergenome.nih.gov/) database [13], meanwhile, the relevant clinical information were also obtained. The gene expression data, which were sequenced on the platform of Affymetrix HT Human Genome U133a microarray, included 568 primary OC tissues, 17 recurrent OC tissues, and 8 adjacent normal tissues. The data is level 3 data downloaded from TCGA, in which the expression level of all probes has been normalized. Based on the annotation platform, probes were then mapped into gene symbols. For multiple probes corresponded to a common gene symbol, their values were averaged and defined as the gene expression value.

Cluster analysis and differential analysis

The variance of gene expression levels for each gene in the samples was calculated, and the gene with variance less than 20% of the total variance of all genes was removed. Meanwhile, the median of gene expression level for each gene in each sample was used as the statistical indicator, and then the gene with median less than 20% of the total median of all genes was eliminated. The expression levels of the genes with potential expression changes in each sample were performed centralization. To pre-classify the samples into two groups, cluster analysis was conducted using the ConsensusClusterPlus package [14] in R. Subsequently, the limma package (Linear Models for Microarray Analysis, http://www.bioconductor.org/packages/release/bioc/html/limma.html) [15] in R was utilized to perform differential analysis for each gene in the pre-classified samples, and the genes with p-value < 0.001 were identified as feature genes.

Screening of stable feature genes and key prognostic genes

To obtain novel sample classification and feature genes, the expression levels of feature genes were iteratively used for the above cluster analysis and differential analysis. Then, the novel sample classification and feature genes were separately compared with the previous sample classification and feature genes. If both of them were inconsistent, the expression matrix of the novel feature genes would be applied for the next iteration. The stable feature genes were obtained until at least one of the sample classification and the feature genes was consistent. To further identify the stable feature genes associated with prognosis, the expression level of each gene in each sample was used to classify the samples according to the average expression level of the gene. Then, Kaplan-Meier (KM) survival analysis [16] was carried out, and the feature genes with p-value < 0.01 were taken as prognostic seed genes.

The prognostic seed genes were performed Euclidean distance cluster to reclassify the samples. Then, the reclassified samples were conducted KM survival analysis [16]. Based on this sample classification, the differential expression level of each prognostic seed gene was calculated using the limma package [15] in R, with p-value < 0.001 as the threshold. Log2 fold change (FC) value (with fixed interval) was used as the cut-off criterion for screening gene set. Then, cluster analysis and KM survival analysis [16] successively were performed, and the most significant gen were considered as the ultimately key prognostic genes.

Functional enrichment analysis

Gene Ontology (GO, http://www.geneontology.org/) aims to provide functions of genes and gene products from the following aspects: biological process (BP), molecular function (MF), and cellular component (CC) [17]. Using the clusterProfiler package (http://bioconductor.org/packages/release/bioc/html/clusterProfiler.html) [18] in R, the key prognostic genes were performed GO functional enrichment analysis. The terms with q-value < 0.05 were selected as the significant functions.

Multivariate survival analysis

To examine the overall influence of the key prognostic genes on prognosis, multivariate survival analysis was conducted for the key prognostic genes. Besides, Receiver Operating Characteristic (ROC) curve was drawn using the survivalROC package [19] in R.

Validation of the key prognostic genes using an independent microarray data

To confirm that the repeatability and portability of key prognostic genes, microarray data of GSE17260 were downloaded from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo/) and used for survival analysis. GSE17260, which was sequenced on the platform of Agilent-014850 Whole Human Genome Microarray 4x44K G4112F, included a total of 110 serous OC samples. Using the survival package [20] in R, cox regression analysis was conducted for the microarray data.

Results

Data preprocessing, cluster analysis and differential analysis

Through data preprocessing, the expression values of a total of 12042 genes were acquired. After screening the primary OC tissues with clinical information, a total of 564 samples were obtained. The variance and median of gene expression levels were calculated, and then the genes with little expression changes among the samples were removed. After that, a total of 8873 genes were screened. The expression levels of the 8873 genes in the 564 samples were performed centralization. Then, the 564 samples were pre-classified into two groups (one group had 195 samples and the other group had 369 samples) through cluster analysis to identify the prognosis difference among all samples. The heatmap of cluster analysis showed that the 8873 genes could distinguish different samples (Fig. 1). Subsequently, a total of 3668 genes were identified as feature genes using differential analysis.

Fig. 1
figure 1

The cluster heatmap for the 8873 genes

Screening of stable feature genes and key prognostic genes

Through the loop iteration of the expression levels of feature genes, a total of 3393 stable feature genes were obtained. The two clusters with different prognosis status of the stable feature genes included 211 samples and 353 samples, respectively. The samples under the same cluster exhibited high correlation (Fig. 2). The clinical features of the two sample groups were further observed, and the result showed that the two group (cluster 1 and cluster 2) samples had significant differences in both the stage (Table 1A) and the grade (Table 1B).

Fig. 2
figure 2

The cluster heatmap for the 3393 stable feature genes

Table 1 The stage and grade distribution of the two groups of samples divided by the 3393 stable feature genes

Using KM survival analysis, a total of 75 prognostic seed genes were identified. Afterwards, cluster analysis was conducted for the prognostic seed genes, and the cluster heatmap suggested that the samples could be obviously divided into two groups (one group had 479 samples and the other group had 85 samples) (Fig. 3). Using log-rank test, the survival analysis for the two group samples showed that they had significant differences in prognosis (Fig. 4), indicating that the prognostic seed genes could classify the samples in prognostic level.

Fig. 3
figure 3

The cluster heatmap for the 75 prognostic seed genes

Fig. 4
figure 4

The result of survival analysis for the two groups of samples divided by the 75 prognostic seed genes

The cluster heatmap showed that the expression levels of some genes among the 75 prognostic seed genes were not very obvious, thus the key prognostic genes were further extracted. After trying different log2 FC value for screening gene set, we found that |log2FC| > 0.7 was the most optimal threshold. Under |log2FC| > 0.7, the gene set containing 25 genes had the most significant influence on prognosis, as they can distinguish patients with different survival status. Thus, the 25 genes were selected as the key prognostic genes, including AXL receptor tyrosine kinase (AXL), FBJ murine osteosarcoma viral oncogene homolog (FOS), Kruppel-like factor 6 (KLF6), WD repeat domain 77 (WDR77), dual specificity phosphatase 1 (DUSP1), growth arrest and DNA damage inducible beta (GADD45B), and slit guidance ligand 3 (SLIT3) (Table 2).

Table 2 The information of the 25 key prognostic genes

Functional enrichment analysis

With q-value < 0.05 as the threshold, the 25 key prognostic genes were significantly enriched in 14 terms. The enriched functions mainly included ovulation cycle (q-value = 0.004227, which involved AXL and SLIT3), reproductive structure development (q-value = 0.004227), and regulation of reproductive process (q-value = 0.004227) (Table 3).

Table 3 The GO (Gene Ontology) functions enriched for the 25 key prognostic genes

Multivariate survival analysis

The cluster heatmap of the 25 key prognostic genes suggested that these genes could divide the samples into two groups (Fig. 5). The multivariate survival analysis for the key prognostic genes showed that the overall survival time of the patients in the two groups had significant difference (p-value = 0.00226) (Fig. 6). Therefore, the 25 key prognostic genes could effectively differentiate the samples and were significantly associated with prognosis.

Fig. 5
figure 5

The cluster heatmap for the 25 key prognostic genes

Fig. 6
figure 6

The Kaplan-Meier (KM) survival curve for the two groups of samples divided by the 25 key prognostic genes

Validation of the key prognostic genes using an independent microarray data

The microarray data of GSE17260 were taken as validation dataset to confirm that the repeatability and portability of the key prognostic genes. Multivariate survival analysis showed that the 25 key prognostic genes also had good classification effects for the validation dataset (Fig. 7). This suggested that the 25 key prognostic genes were key genes affecting the prognosis of OC.

Fig. 7
figure 7

The Receiver Operating Characteristic (ROC) curve (a) and Kaplan-Meier (KM) survival curve (b) of multivariate survival analysis for the validation dataset. The results showed that the 25 key prognostic genes also had good classification effects for the validation dataset

Discussion

In this study, a total of 564 samples were obtained from data preprocessing, which were pre-classified into two groups. Afterwards, differential analysis identified 3668 feature genes. Besides, 3393 stable feature genes were obtained through loop iteration, and 75 genes among them were identified as prognostic seed genes. Moreover, 25 prognostic seed genes were selected as the key prognostic genes (including AXL, FOS, KLF6, WDR77, DUSP1, GADD45B, and SLIT3). Multivariate survival analysis indicated that the 25 key prognostic genes could effectively differentiate the samples and were significantly associated with prognosis. In addition, the microarray data of GSE17260 further confirmed that the key prognostic genes were key genes affecting the prognosis of OC.

GADD45 proteins mediate many cellular functions such as cell cycle control, genotoxic stress, DNA repair, and senescence, additionally, GADD45 proteins functions as tumor suppressors through their pro-apoptotic activities [21, 22]. Overexpression of GADD45A may be implicated in the pro-apoptosis effect of the synthetic retinoid CD437 on ovarian cancer cells [23]. Oliveira-Ferrer et al. deem that c-FOS may affect OC progression through exerting pro-apoptotic effect and altering peritoneal adhesion of OC cells [24]. Mahner et al. find that down-regulated c-Fos plays a role in tumor progression in OC and c-Fos may be used as prognostic factor for the disease [25]. Furthermore, KLF6 and its alternative splicing isoform KLF6-SV1 are related to the main clinical characteristics of EOC, thus they may serve as therapeutic targets for changing the development and dissemination of OC [26]. Above evidence declared that GADD45B, FOS, and KLF6 might be correlated with the prognosis of OC.

Ligr et al. find that p44/Mep50/WDR77 is associated with hormone effects during ovarian tumorigenesis [27]. Via regulating the p38 MAPK-mediated p-glycoprotein overexpression, DUSP1 may cause the resistance of human OC cells to paclitaxel [28, 29]. The glucocorticoid administration to OC patients is correlated with increased expression of map kinase phosphatase 1 (MKP1, also known as DUSP1) and serum and glucocorticoid-regulated kinase 1 (SGK1), indicating that glucocorticoids may weaken chemotherapy effect in OC patients by promoting the expression of anti-apoptotic genes [30]. Additionally, MKP1 can be induced by cisplatin via ERK signaling-associated phosphorylation, and the ERK-MKP1 signaling functions in overcoming cisplatin resistance in OC patients [31]. Thus, WDR77 and DUSP1 might play roles in the development of OC.

Previous study find that Growth arrest-specific gene 6 (GAS6)/AXL pathway has an influence on the complex events occurring during the early stage of OC [32]. GAS6/AXL targeting can be an effective mean for inhibiting the progression of metastatic OC, and the soluble AXL receptor is a promising agent for treating the disease [33]. Since cortisol suppressing SLIT/Roundabout (ROBO) pathway contributes to the regeneration of ovarian surface epithelium, the pathway may be a target for controlling the SLIT/ROBO system in OC [34, 35]. Qiu et al. demonstrate that SLIT2 can serve as tumor suppressor in OC, thus it may be used as a promising therapeutic target for the disease [36]. Functional enrichment analysis showed that AXL and SLIT3 were enriched in ovulation cycle, suggesting that AXL and SLIT3 might also be involved in OC through affecting ovulation cycle.

Conclusions

In conclusion, a total of 3668 feature genes and 25 key prognostic genes were screened by bioinformatics analysis. Besides, several key genes (AXL, FOS, KLF6, WDR77, DUSP1, GADD45B, and SLIT3) might be associated with the prognosis of OC. However, the functions of these key genes need to be confirmed by experimental researches in future.

Abbreviations

AXL:

AXL receptor tyrosine kinase

BP:

Biological process

CC:

Cellular component

CXCR4:

Chemokine receptor 4

DUSP1:

Dual specificity phosphatase 1

EOC:

Epithelial ovarian cancer

FC:

Fold change

FOS:

FBJ murine osteosarcoma viral oncogene homolog

FOXM1:

Forkhead box M1

GADD45B:

Growth arrest and DNA damage inducible beta

GAS6:

Growth arrest-specific gene 6

GEO:

Gene Expression Omnibus

GO:

Gene Ontology

HMGA2:

High mobility group A2

KLF6:

Kruppel-like factor 6

KM:

Kaplan-Meier

MF:

Molecular function

MKP1:

Map kinase phosphatase 1

OC:

Ovarian cancer

ROC:

Receiver Operating Characteristic

SGK1:

Glucocorticoid-regulated kinase 1

SLIT3:

Slit guidance ligand 3

TCGA:

The Cancer Genome Atlas

TEAD4:

TEA domain family member 4

WDR77:

WD repeat domain 77

YAP:

Yes-associated protein 1

References

  1. Modugno F, Edwards RP. Ovarian cancer: prevention, detection, and treatment of the disease and its recurrence. Molecular mechanisms and personalized medicine meeting report. Int J Gynecol Cancer. 2012;22(8):45–57.

    Article  Google Scholar 

  2. Cancer UK. Treating advanced ovarian cancer. 2014.

    Google Scholar 

  3. Mcguire S. World Cancer Report 2014. Geneva, Switzerland: World Health Organization, International Agency for Research on Cancer, WHO Press, 2015. Advances in Nutrition. 2016;7(2):418–9.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Jayson GC, Kohn EC, Kitchener HC, Ledermann JA. Ovarian cancer. Lancet. 2014;384(9951):1376–88.

    Article  PubMed  Google Scholar 

  5. Li J, Jiang K, Qiu X, et al. Overexpression of CXCR4 is significantly associated with cisplatin-based chemotherapy resistance and can be a prognostic factor in epithelial ovarian cancer. BMB Rep. 2014;47(1):33–8.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Xia Y, Chang T, Wang Y, et al. YAP Promotes Ovarian Cancer Cell Tumorigenesis and Is Indicative of a Poor Prognosis for Ovarian Cancer Patients. Plos One. 2014;9(3):e91770.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Tan G, Cao X, Dai Q, et al. A novel role for microRNA-129-5p in inhibiting ovarian cancer cell proliferation and survival via direct suppression of transcriptional co-activators YAP and TAZ. Oncotarget. 2015;6(11):8676–86.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Cai M, Hu Z, Liu J, et al. Beclin 1 expression in ovarian tissues and its effects on ovarian cancer prognosis. Int J Mol Sci. 2014;15(4):5292–303.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Zhao Y, Chen S, Gou WF, Xiao LJ, Takano Y, Zheng HC. Aberrant Beclin 1 expression is closely linked to carcinogenesis, differentiation, progression, and prognosis of ovarian epithelial carcinoma. Tumor Biology. 2014;35(3):1955–64.

    Article  CAS  PubMed  Google Scholar 

  10. Califano D, Pignata S, Losito NS, et al. High HMGA2 Expression and High Body Mass Index Negatively Affect the Prognosis of Patients With Ovarian Cancer. J Cell Physiol. 2014;229(1):53–9.

    CAS  PubMed  Google Scholar 

  11. Ning W, Yu W, Li W, et al. Overexpression of FOXM1 predicts poor prognosis and promotes cancer cell proliferation, migration and invasion in epithelial ovarian cancer. J Transl Med. 2014;12(1):1–13.

    Article  Google Scholar 

  12. Zhao F, Siu MK, Jiang L, et al. Overexpression of forkhead box protein M1 (FOXM1) in ovarian cancer correlates with poor patient survival and contributes to paclitaxel resistance. Plos One. 2014;9(11):e113478.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Network CGAR. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519–25.

    Article  Google Scholar 

  14. Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26(12):1572–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ritchie ME, Phipson B, Wu D, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.

  16. Lacny S, Wilson T, Clement F, et al. Kaplan-Meier Survival Analysis Overestimates the Risk of Revision Arthroplasty: A Meta-analysis. Clin Orthop Relat Res. 2015;473(11):3431–42.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Carbon S, Ireland A, Mungall CJ, et al. AmiGO: online access to ontology and annotation data. Bioinformatics. 2009;25(2):288–9.

    Article  CAS  PubMed  Google Scholar 

  18. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics A Journal of Integrative Biology. 2012;16(5):284–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Heagerty PJ, Thomas L, Pepe MS. Time-dependent ROC curves for censored survival data and a diagnostic marker. Biometrics. 2000;56(2):337–44.

    Article  CAS  PubMed  Google Scholar 

  20. Akira M, Yuya M, Yoshihisa N, et al. Kaplan–Meier survival analysis and Cox regression analyses regarding right ventricular septal pacing: Data from Japanese pacemaker cohort. Data Brief. 2016;8:1303–7.

    Article  Google Scholar 

  21. Tamura RE, de Vasconcellos JF, Sarkar D, Libermann TA, Fisher PB, Zerbini LF. GADD45 proteins: central players in tumorigenesis. Curr Mol Med. 2012;12(5):634–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Salvador JM, Brown-Clay JD, Fornace AJ. Gadd45 in Stress Signaling, Cell Cycle Control, and Apoptosis. Adv Exp Med Biol. 2013;793(10):1–19.

    Article  CAS  PubMed  Google Scholar 

  23. Jiang T, Soprano DR, Soprano KJ. GADD45A is a mediator of CD437 induced apoptosis in ovarian carcinoma cells. J Cell Physiol. 2007;212(3):771–9.

    Article  CAS  PubMed  Google Scholar 

  24. Oliveira-Ferrer L, Rößler K, Haustein V, et al. c-FOS suppresses ovarian cancer progression by changing adhesion. Br J Cancer. 2013;110(3):753–63.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mahner S, Baasch C, Schwarz J, et al. C-Fos expression is a molecular predictor of progression and survival in epithelial ovarian carcinoma. Br J Cancer. 2008;99(8):1269–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Difeo A, Narla G, Hirshfeld J, et al. Roles of KLF6 and KLF6-SV1 in ovarian cancer progression and intraperitoneal dissemination. Clin Cancer Res. 2006;12(12):3730–9.

    Article  CAS  PubMed  Google Scholar 

  27. Ligr M, Patwa RR, Daniels G, et al. Expression and function of androgen receptor coactivator p44/Mep50/WDR77 in ovarian cancer. Plos One. 2011;6(10):e26250.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kang YS, Seok HJ, Jeong EJ, et al. DUSP1 induces paclitaxel resistance through the regulation of p-glycoprotein expression in human ovarian cancer cells. Biochem Biophys Res Commun. 2016;478(1):403–9.

    Article  CAS  PubMed  Google Scholar 

  29. Kang Y, Nagaraja AS, Armaizpena GN, et al. Adrenergic stimulation of DUSP1 impairs chemotherapy response in ovarian cancer. Clin Cancer Res. 2015;22(7):1713–24.

  30. Melhem A, Yamada SD, Fleming GF, et al. Administration of glucocorticoids to ovarian cancer patients is associated with expression of the anti-apoptotic genes SGK1 and MKP1/DUSP1 in ovarian tissues. Clin Cancer Res. 2009;15(9):3196–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang J, Zhou JY, Wu GS. ERK-dependent MKP-1-mediated cisplatin resistance in human ovarian cancer cells. Cancer Res. 2007;67(24):11933–41.

    Article  CAS  PubMed  Google Scholar 

  32. Sun W, Fujimoto J, Tamaya T. Coexpression of Gas6/Axl in Human Ovarian Cancers. Oncology. 2004;66(6):450–7.

    Article  CAS  PubMed  Google Scholar 

  33. Rankin EB, Fuh KCTaylor TE, Krieg AJ, et al. AXL is an essential factor and therapeutic target for metastatic ovarian cancer. Cancer Res. 2010;70(19):7570–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dickinson RE, Fegan KS, Ren X, Hillier SG, Duncan WC. Glucocorticoid Regulation of SLIT/ROBO Tumour Suppressor Genes in the Ovarian Surface Epithelium and Ovarian Cancer Cells. Plos One. 2011;6(11):e27792.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dickinson RE, Myers M, Duncan WC. Novel regulated expression of the SLIT/ROBO pathway in the ovary: possible role during luteolysis in women. Endocrinology. 2008;149(10):5024–34.

    Article  CAS  PubMed  Google Scholar 

  36. Qiu H, Zhu J, Yu J, Pu H, Dong R. SLIT2 is epigenetically silenced in ovarian cancers and suppresses growth when activated. Asian Pac J Cancer Prev. 2011;12(3):791–5.

    PubMed  Google Scholar 

Download references

Acknowledgements

This study was supported by the national natural science fund projects (grant number: 81571387) and Funds for start-up program in Shanghai Changhai Hospital (grant number: CH201506).

Funding

none.

Availability of data and materials

The raw data were collected and analyzed by the Authors, and are not ready to share their data because the data have not been published.

Authors’ contributions

JJZ and LL participated in the design of this study.LL,SNL HF and SYC performed the statistical analysis. LL carried out the study and collected important background information.LL,SYC and JJZ drafted the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interest.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Publisher’s Note

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

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Junjie Zhang.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, L., Cai, S., Liu, S. et al. Bioinformatics analysis to screen the key prognostic genes in ovarian cancer. J Ovarian Res 10, 27 (2017). https://doi.org/10.1186/s13048-017-0323-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13048-017-0323-6

Keywords