Open Access

The core genes involved in the promotion of depression in patients with ovarian cancer

  • Authors:
    • Yuexiong Yi
    • Yanyan Liu
    • Kejia Wu
    • Wanrong Wu
    • Wei Zhang
  • View Affiliations

  • Published online on: September 30, 2019     https://doi.org/10.3892/ol.2019.10934
  • Pages: 5995-6007
  • Copyright: © Yi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to identify the core genes and pathways involved in depression in patients with ovarian cancer (OC) who suffer from high or low‑grade depression. The dataset GSE9116 from Gene Expression Omnibus database was analyzed to identify differentially expressed genes (DEGs) in these patients. To elucidate how certain genes could promote depression in patients with OC, pathway crosstalk, protein‑protein interaction (PPI) and comprehensive gene‑pathway analyses were determined using WebGestalt, ToppGene and Search Tool for the Retrieval of Interacting Genes and gene ontology analysis. Key genes and pathways were extracted from the gene‑pathway network, and gene expression and survival analysis were evaluated. A total of 93 DEGs were identified from GSE9116 dataset, including 84 upregulated genes and nine downregulated genes. The PPI, pathway crosstalk and comprehensive gene‑pathway analyses highlighted C‑C motif chemokine ligand 2 (CCL2), Fos proto‑oncogene, AP‑1 transcription factor subunit (FOS), serpin family E member 1 (SERPINE1) and serpin family G member 1 (SERPING1) as core genes involved in the promotion of depression in patients with OC. These core genes were involved in the following four pathways ‘Ensemble of genes encoding ECM‑associated proteins including ECM‑affiliated proteins’, ‘ECM regulators and secreted factors’, ‘Ensemble of genes encoding extracellular matrix and extracellular matrix‑associated proteins’ and ‘MAPK signaling pathway and IL‑17 signaling pathway’. The results from gene expression and survival analysis demonstrated that these four key genes were upregulated in patients with OC and high‑grade depression and could worsen patients' survival. These results suggested that CCL2, FOS, SERPINE1 and SERPING1 may serve a crucial role in the promotion of depression in patients with OC. This finding may provide novel markers for predicting and treating depression in patients with OC; however, the underlying mechanisms remain unknown and require further investigation.

Introduction

Ovarian cancer (OC) is a fatal malignancy of the female reproductive system (1). In 2018, there were 22,240 new cases of ovarian malignancies, and 14,070 OC-associated mortality cases (2). Furthermore, the incidence and mortality rates of malignant ovarian tumors in China reached 52.1/100,000 and 22.5/100,000, respectively, in 2015 (3). Due to the non-specific symptoms of OC, 70% of Chinese patients are diagnosed with a later stage of the disease at first diagnosis (4).

Previous studies demonstrated that ~38% of patients with cancer display major emotional distress, including anxiety and depression (57). In patients with OC, the incidence of depression and anxiety is of 82 and 92%, respectively (8). In addition, the degree of depression or anxiety is highly variable in patients with OC (9).

At present, the majority of studies have focused on how depression affects the prognosis of OC (1012); however, the pathogenesis of depression in OC remains poorly investigated, particularly at a molecular level. By using a miRNA-mRNA regulation network, Wu et al (13) identified 12 miRNA-mRNAs pairs (miR-629-5p-FGF1, miR-629-5p-AKT3, miR-629-5p-MAGI2, miR-933-BDNF, miR-933-MEF2A, miR-23b-3p-TJP1, miR-23b-3p-JMJD1, miR-23b-3p-APAF1, miR-23b-3p-CAB39, miR-1265-CDKN1B, miR-33b-3p-CDKN1B and miR-33b-3p-F2R) that could be associated with the development of major depressive disorder (MDD) in patients with OC. Furthermore, Rahman et al (14) identified 34 differentially expressed genes (DEGs) associated with depression in patients with OC and demonstrated that CXCL12, ARL4C, NQO2 were associated with worse survival in patients with OC. As depressed patients display higher mortality rates (15) and since a different mental status can lead to different clinical outcomes (16) and prognosis (17), it is crucial to further understand the molecular mechanisms underpinning the onset of depression in patients with OC.

The present study aimed to identify DEGs in patients with OC and high or low-grade depression using bioinformatics analyses, and to determine the potential hub genes and pathways that may serve critical roles in the onset of depression in patients with OC.

Materials and methods

Study design

The flowchart for this study is presented in Fig. 1. DEGs were first identified in GSE9116 dataset (18,19) according to the following criteria: P<0.05 and absolute log2 value of fold change >1 (|log2(FC)|>1). The top three up- and downregulated DEGs were collected as potential key genes for further analysis. Subsequently, Gene Ontology (GO) (https://www.webgestalt.org/) and pathways enrichment analyses (https://toppgene.cchmc.org/) were performed using WebGestalt (20) and ToppGene (21) tools separately, and the protein-protein interaction (PPI) and pathway crosstalk networks were constructed. The gene-pathway network was constructed by mapping the hub genes into hub pathways extracted from PPI and pathway crosstalk network according to the criterion of nodal degree > average. Subsequently, the core genes with pathways were extracted from the gene-pathway network according to the criterion node degree > average. Eventually, the core genes and the top three up- and downregulated DEGs, which were defined as key genes, were studied by expression and survival analysis.

Microarray dataset

The GSE9116 dataset describing the gene expression profile of OC (18,19) and established on the platform of GPL96 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GPL96), was downloaded from Gene Expression Omnibus (GEO) database (22). To the best of our knowledge, this dataset is the only one that studied the impact of depression on gene expression in patients with OC. This dataset contains ten samples of primary OC (grade 3 serous cancer or serous adenocarcinoma), including five samples from patients with low-grade depression (LD) and five samples from patients with high-grade depression (HD). The samples and psychosocial data were collected from patients who were diagnosed with ovarian epithelial cancer, peritoneal cancer, or cancer of the fallopian tube, and who underwent primary surgical resection of ovarian carcinoma (14). A patient with a Center for Epidemiologic Studies Depression Scale score (23) ≥16 and a Social Provisions Scale-Attachment score (24) ≤15 was defined as having high-grade depression. Otherwise, patients were defined as having low-grade depression (18).

Identification of DEGs

The GSE9116 dataset was divided into two groups: HD and LD groups. Genes were considered differentially expressed between HD and LD groups if they exhibited a |log2(FC)|>1 and P<0.05 calculated using GEO2R (16) with the limma package (25). The top three up- and downregulated DEGs were considered as potential key genes for further analysis.

GO enrichment analysis

GO analysis was performed using WebGestalt tool (20) on the DEGs. Enriched biological terms for cellular components (CC), biological process (BP) and molecular functions (MF) with a P<0.05 were identified.

PPI network and identification of hub genes

The Search Tool for the Retrieval of Interacting Genes (STRING) (26) database and Cytoscape (version 3.5.1) (27) were used to identify hub genes. The PPI network was searched by gene symbols and the minimum required interaction score was set at 0.7 to ensure high confidence in the results. Nodes that were not connected to the major network were removed to reduce the error detection rate. The CentiScaPe plug-in (28) was used to calculate the degree of each node. Nodes were considered as hub if their degree was larger than the average.

Pathway enrichment and crosstalk analysis

For pathway enrichment, DEGs were mapped using the Kyoto Gene and Genome Encyclopedia (KEGG) database (29) and BioCarta (30) using the online analysis tool (21). Pathways with a P<0.05 were considered significant.

Pathway crosstalk analysis was conducted on the pathways defined above based on the assumption that two pathways are considered crosstalking if they share a proportion of genes (31,32). Briefly, pathways containing <3 genes were excluded as pathways with too few genes may contain insufficient biological information. Two indicators named Jaccard Coefficient (33) and Overlapping Coefficient (34), were used to measure the overlap between two pathways and were calculated as follows: Jaccard Coefficient = |(A∩B)/(A∪B) and Overlapping Coefficient = [(|A∩B|)/(min(|A|,|B|)], where A and B represent the list of genes in the two pathways, and min indicated the minimum number of genes between |A| and |B|. Subsequently, pairs of pathways with more than one overlapping gene were retained. Once the pathway crosstalking network was obtained, a subnetwork representing the hub pathways was identified according to the criterion nodal degree > average.

Comprehensive gene-pathway analysis

By mapping hub genes into the subnetwork of pathway crosstalk using KEGG (29) and BioCarta (30), a comprehensive gene-pathway network was obtained to further investigate the association between genes and pathways. The core genes and pathways in the gene-pathway network were identified according to the criterion nodal degree > average.

Expression and survival analysis of key genes

The expression of core genes in the sub gene-pathway network and of the top three up and downregulated DEGs were extracted from the GSE9116 dataset and evaluated for regulations. Furthermore, to analyze the effect of the key genes on patients' overall survival (OS), progression-free survival (PFS) and post-progression survival (PPS), patients were stratified according to the expression of the key genes and evaluated using Kaplan-Meier analysis (35). Kaplan-Meier analysis provides a survival assessment of prognosis-related genes in OC patients whose data obtained from The Cancer Genome Atlas (36) and 14 GSE profiles [GSE14764 (37), GSE15622 (38), GSE18520 (39), GSE19829 (40), GSE23554 (41), GSE26193 (42,43), GSE26712 (44,45), GSE27651 (46), GSE30161 (47), GSE3149 (48), GSE51373 (49), GSE63885 (50), GSE65986 (51) and GSE9891 (52)]. Patients were grouped by median gene expression and hazard ratio (HR) with 95% confidence intervals (CI) and log-rank P-values were calculated. P<0.05 was considered to indicate a significant difference.

Results

Identification of DEGs

According to the criteria P<0.05 and |Log2(FC)|>1, 93 DEGs were identified when comparing the HD and LD groups. A total of 84 DEGs were upregulated and nine were downregulated. Plasminogen activator, tissue type (PLAT), activating transcription factor 3 (ATF3) and cellular communication network factor 2 (CTGF) were the top three upregulated genes with the highest change in expression. Heat shock 70 kDa protein 1B (HSPA1B), endonuclease G (ENDOG) and EPS8 like 1 (EPS8L1) were the top three downregulated genes. A heatmap with logFC values of the 93 DEGs is presented in Fig. 2.

GO enrichment analysis

GO enrichment analysis was performed on the 93 DEGs using WebGestalt (Fig. 3). In terms of BP, DEGs were primarily involved in ‘biological regulation’, ‘metabolic process’, ‘multicellular organismal process’ and ‘response to stimulus’. In terms of CC, DEGs were enriched in ‘membrane’, ‘nucleus’, ‘endomembrane system’ and ‘vesicle’. In terms of MF, DEGs mainly participated in ‘protein binding’, ‘ion binding’ and ‘hydrolase activity’.

Construction of the PPI network and identification of hub genes

Using DEGs and the STRING database, a PPI network containing 34 nodes and 55 edges was obtained (Fig. 4A). The degree of each node was calculated using CentiScaPe (Table I). A total of 16 hub genes of which degree > average were extracted from the PPI network (Fig. 4B). This subnetwork was divided into two clusters that contained eight and seven genes, respectively. These two clusters were connected by C-C motif chemokine ligand 2 (CCL2) (Fig 4B).

Table I.

Characteristics of genes identified from protein-protein interaction network.

Table I.

Characteristics of genes identified from protein-protein interaction network.

RankGeneDegree of each node
Overall average value 3.2352
  1FOS9
  2JUNB7
  3DUSP16
  4EGR16
  5FOSB6
  6SERPINE16
  7SPARC5
  8TIMP35
  9JUND5
10SEPP14
11SERPING14
12ZFP364
13ATF34
14COL3A14
15DCN4
16CCL24
17LUM3
18CTGF2
19CXCL122
20CTSB2
21HLA-DPA12
22CHPT12
23PLA2G72
24PLA2G162
25SMARCA41
26CST31
27KLC11
28PNOC1
29TMEM176A1
30TMEM176B1
31PRELP1
32DUSP61
33PLOD21
34PLAT1
Pathway enrichment and crosstalk analysis

Pathway enrichment analysis was performed using ToppGene database. The results indicated that the top five significantly enriched pathways were ‘Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins’, ‘Ensemble of genes encoding core extracellular matrix including ECM glycoproteins, collagens and proteoglycans’, ‘Complement and coagulation cascades’, ‘Pertussis toxin-insensitive CCR5 Signaling in Macrophage’ and ‘Rheumatoid arthritis’ (Table II).

Table II.

Results from pathway enrichment analysis.

Table II.

Results from pathway enrichment analysis.

PathwaysSourceP-valueGenes in the pathway
Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteinsBIOCARTA 1.19×10−08COL3A1, CRISPLD2, FBLN5, SPARC, MXRA5, DCN, EGFL6, TIMP3, PLXNC1, PRELP, SERPINE1, XCL2, CST3, SERPING1, CCL2, PLAT, CTGF, LUM, CTSB, XCL1, PLOD2, CXCL12
Ensemble of genes encoding core extracellular matrix including ECM glycoproteins, collagens and proteoglycansBIOCARTA 1.73×10−05COL3A1, CRISPLD2, FBLN5, SPARC, MXRA5, DCN, PRELP, CTGF, LUM
Complement and coagulation cascadesKEGG 6.90×10−05SERPINE1, SERPING1, PLAT, C4A, C4B
Pertussis toxin-insensitive CCR5 signaling in MacrophageBIOCARTA 1.20×10−04FOS, CCL2, CXCL12
Rheumatoid arthritisKEGG 1.29×10−04ATP6V1B1, HLA-DPA1, FOS, CCL2, CXCL12
Ensemble of genes encoding ECM-associated proteins including ECM-affiliated proteins, ECM regulators and secreted factorsBIOCARTA 1.85×10−04EGFL6, TIMP3, PLXNC1, SERPINE1, XCL2, CST3, SERPING1, CCL2, PLAT, CTSB, XCL1, PLOD2, CXCL12
Genes encoding enzymes and their regulators involved in the remodeling of the extracellular matrixBIOCARTA 3.07×10−04TIMP3, SERPINE1, CST3, SERPING1, PLAT, CTSB, PLOD2
MAPK signaling pathwayKEGG 4.66×10−04FOS, DUSP1, DUSP6, JUND, FGFR3, HSPA1A, HSPA1B
PertussisKEGG 7.77×10−04FOS, SERPING1, C4A, C4B
Antigen processing and presentationKEGG 8.16×10−04HLA-DPA1, CTSB, HSPA1A, HSPA1B
Genes encoding proteoglycansBIOCARTA 9.00×10−04DCN, PRELP, LUM
IL-17 signaling pathwayKEGG 1.65×10−03FOS, FOSB, JUND, CCL2
Lectin Induced Complement PathwayBIOCARTA 1.87×10−03C4A, C4B
Fibrinolysis PathwayBIOCARTA 1.87×10−03SERPINE1, PLAT
Ether lipid metabolismKEGG 1.88×10−03PLA2G7, PLA2G16, CHPT1
Estrogen signaling pathwayKEGG 2.00×10−03FOS, HSPA1A, HSPA1B, GABBR1
AGE-RAGE signaling pathway in diabetic complicationsKEGG 2.08×10−03COL3A1, EGR1, SERPINE1, CCL2
Classical Complement PathwayBIOCARTA 2.56×10−03C4A, C4B
Platelet Amyloid Precursor Protein PathwayBIOCARTA 2.56×10−03SERPINE1, PLAT
B Cell Survival PathwayBIOCARTA 3.36×10−03FOS, JUND
Staphylococcus aureus infectionKEGG 3.51×10−03HLA-DPA1, C4A, C4B
Genes encoding structural ECM glycoproteinsBIOCARTA 4.29×10−03CRISPLD2, FBLN5, SPARC, MXRA5, CTGF
Complement PathwayBIOCARTA 4.73×10−03C4A, C4B
Osteoclast differentiationKEGG 5.52×10−03FOS, FOSB, JUNB, JUND

For the pathway crosstalk analysis, 17 out of 24 pathways that contained ≥2 genes met the crosstalk analysis criteria and were selected to construct the network (Fig. 4C). The thickness of the edges indicated the average values of Jaccard and the Overlapping Coefficient that represented the overlapping level of genes between two pathways. By selecting the nodes with degree > average, a subnetwork of pathway crosstalk with 7 nodes and 11 edges was constructed (Fig. 4D).

Comprehensive gene-pathway analysis

After mapping the hub genes onto the subnetwork of pathways provided by KEGG and BioCarta, a potential gene-pathway network including 7 essential pathways and 11 hub genes was constructed (Fig. 4E). The results from this network demonstrated that CCL2 and Fos proto-oncogene, AP-1 transcription factor subunit (FOS) participated in most of the pathways. Based on the number of genes involved, the top three pathways were ‘Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins’, ‘Ensemble of genes encoding ECM-associated proteins including ECM-affiliated proteins, ECM regulators and secreted factors’ and ‘IL-17 signaling pathway’ (Fig. 4E).

To identify the main nodes (including genes and pathways) in the gene-pathway network, nodes with degree > average were selected (Fig. 4F). The results demonstrated that CCL2, FOS, serpin family G member 1 (SERPING1) and serpin family E member 1 (SERPINE1) and the four pathways ‘Ensemble of genes encoding ECM-associated proteins including ECM-affiliated proteins, ECM regulators and secreted factors’, ‘Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins’, ‘MAPK signaling pathway’ and ‘IL-17 signaling pathway’ were identified and may serve a crucial role in the development of depression in patients with OC.

Association between key genes and the six top DEGs

The evaluation of the interaction between the four key genes (CCL2, FOS, SERPING1 and SERPINE1) and the six top DEGs (upregulated; PLAT, ATF3 and CTGF and downregulated; HSPA1B, ENDOG and EPS8L1) in the PPI (Fig 4A) demonstrated that HSPA1B, ENDOG, EPS8L1 were not found since they were not connected to the major network. Furthermore, PLAT was interplayed with SERPINE1, CTGF interacted with CCL2 and ATF3 was connected to FOS.

Expression and survival analysis of key genes

As presented in Fig. 5, all core genes and top three upregulated DEGs were significantly upregulated in the HD group, which was not the case for the top three downregulated DEGs compared with the LD group.

The results from survival analysis indicated that the top 3 upregulated DEGs had similar effects as the core genes, which was not the case for the top 3 downregulated DEGs (Fig. 6). In particular, higher expression of ATF3 (HR, 1.19; 95% CI, 1.03–1.37; P=0.019) and SERPINE1 (HR, 1.27; 95% CI, 1.11–1.46; P=0.00056) and lower expression of ENDOG (HR, 0.83; 95% CI, 0.73–0.95; P=0.0061) and EPS8L1 (HR, 0.82; 95% CI, 0.72–0.94; P=0.004) were significantly associated with worse OS. Furthermore, ATF3 (HR, 1.19; 95% CI, 1.03–1.36; P=0.016), CTGF (HR, 1.28; 95% CI, 1.12–1.47; P=0.00033), HSPA1B (HR, 1.15; 95% CI, 1.01–1.52; P=0.04), EPS8L1 (HR, 1.17; 95% CI, 1.12–1.34; P=0.0021) and all core genes, including CCL2 (HR, 1.24; 95% CI, 1.09–1.41; P=0.0013), FOS (HR: 1.21, 95%CI: 1.06–1.37; P=0.0048), SERPINE1 (HR, 1.38, 95% CI, 1.21–1.57; P=1.1e-06) and SERPING1 (HR, 1.22; 95% CI, 1.05–1.41; P=0.0084), were significantly associated with a lower PFS. However, lower expression of HSPA1B (HR, 0.67; 95% CI, 0.56–0.8; P=0.000016), CCL2 (HR, 0.83; 95% CI, 0.7–0.99; P=0.038) and SERPING1 (HR, 0.8; 95% CI, 0.67–0.97; P=0.023) and high expression of SERPINE1 (HR, 1.42; 95% CI, 1.18–1.72; P=0.00026) were significantly associated with worse PPS. The other genes had no significant effect on OS, PFS or PPS.

Discussion

The incidence of cancer continues to rise annually (53). With the advancement of psychological research, increasing attention is being paid to the mental health of patients with malignant tumors (54). Depression is a common psychological issue observed in cancer patients. It can reduce the efficacy of treatments, delay the recovery time and reduce the quality of life of patients (55). At similar stages of cancer cell differentiation or treatments, patients with depression may have a worse prognosis compared with non-depressed patients (56). It has been reported that patients with OC whom exhibit a positive attitude have an improved quality of life and prognosis compared with patients with negative emotions (57). It is therefore crucial to determine the underlying molecular mechanisms linking depression with poorer prognosis for patients with ovarian cancer.

Whereas the original study of GSE9116 used a promoter-based bioinformatics strategy to investigate the effect of β-adrenergic signaling (18) and focused on the genetic interaction locus in the human interleukin (IL)-6 promoter (single nucleotide polymorphisms rs1800795) (19), the present study aimed to identify key genes and pathways associated with depression in patients with OC using PPI networks, pathway crosstalk and gene-pathway analyses. The results from this study may provide an improved understanding underlying the link between depression in and a poorer prognosis in patients with OC.

The results from the present study demonstrated that CCL2 may induce depression in patients with OC through ECM-related and IL-17 signaling pathways. Previous studies indicated that high expression of CCL2 is associated with depression. Leighton et al (58) performed a meta-analysis, which included 4688 subjects (765 depressed healthy patients vs. 1528 not depressed healthy patients, and 742 depressed patients with inflammatory illness vs. 1653 not depressed patients with inflammatory illness) and reported that CCL2 blood levels were significantly higher in depressed patients with or without inflammatory illness compared with the control. Another meta-analysis reported similar results for CCL2 (59). In addition, CCL2 was upregulated in cancerous cells, and this required the participation of IL-17 (60), and CCL2 induced tumor cell proliferation and stimulated tumor cell migration and invasion into the surrounding extracellular matrix in order to promote tumor cell intravasation into the circulation (61).

The present study demonstrated an association between FOS expression and depression via MAPK and IL-17 signaling pathways in patients with OC. Yu et al (62) reported that human depression is associated with cognitive deficits, and that FOS is upregulated in cognitively impaired mice. Kung et al (63) demonstrated that FOS expression is higher in preproenkephalin-knockout mice, and induced anxiety and depression-like symptoms of post-traumatic stress disorder compared with wild-type mice. In addition, it was reported that FOS expression is regulated by the MAPK pathway (64,65). Furthermore, IL-17 can mediate inflammatory responses via FOS activation (66) and enhance the recruitment of activated FOS in synergy with IL-6 (67).

The results from the present study indicated that SERPINE1 and SERPING1 could induce depression in patients with OC through ECM-related pathways. Both SERPINE1 and SERPING1 belong to the SERPINs family of serine protease inhibitors that regulate proteases involved in fibrinolysis, coagulation, inflammation, cell mobility, cellular differentiation and apoptosis (68). SERPINE1, which is known as plasminogen activator inhibitor-1 (PAI-1), is described as a major physiological inhibitor of endogenous plasminogen activator which inhibits fibrin degradation, promotes fibrin deposition on blood vessel walls and stimulates smooth muscle cell proliferation (69). Furthermore, SERPINE1 may be involved in the pathogenic and therapeutic mechanisms of MDD. Yamamoto et al (70) reported that stress induced SERPINE1 upregulation in a tissue- and cell type-specific manner. In addition, previous studies demonstrated that both women (71) and men (72) with MDD present higher blood PAI-1 levels compared with healthy subjects. Although these findings were not described in patients with OC, they are similar to the results from the present study. Previous studies indicated that SERPINE1 is a stress-associated gene and that its genetic variants may contribute to the causes of depression and the acute therapeutic response to selective serotonin reuptake inhibitors in MDD (73). However, the promoter polymorphisms of SERPINE1 gene were not associated with Alzheimer's disease-related depression, but they may be associated with the response to antidepressant treatments (74). Therefore, the mechanism by which SERPINE1 induces depression in patients with OC requires further investigation. SERPING1 is also referred to as a complement system C1 inhibitory gene [complement component 1 inhibitor (C1INH)], which is located on chromosome 11q.11-q13.1 on GeneMap (75). SERPING1 has been reported to be associated with plasma protein supplementation and to be a member of the serine protease inhibitor gene family (76). Numerous studies have reported that SERPING1 is downregulated in patients with hereditary angioedema and depression (7779). Furthermore, Ditzen et al (80) demonstrated that SERPING1 expression levels were decreased in the cerebrospinal fluid of depressed patients compared with healthy controls. Since the results from the present study demonstrated that patients with OC and depression exhibited higher SERPING1 expression, SERPING1 may cause depression through other pathways in cancer microenvironment. In addition, SERPINE1 and SERPING1 are associated with extracellular matrix regulation (68). The plasminogen activator/plasmin system serves a crucial role in ECM degradation, and SERPINE1 is a physiological inhibitor of plasminogen activators (81). Furthermore, SERPINE1 upregulation promotes the deposition of ECM components (82), and SERPING1 interacts with extracellular matrix components to inhibit protease activity (83). In addition, interactions between SERPING1 and extracellular matrix components may result in an increase in C1INH concentration at inflammation sites (84). However, as SERPINE1/SERPING1 interaction with ECM remains unclear, further investigation is required.

ATF3 and CTGF upregulation have been demonstrated to be associated with depression. Green et al (85) reported that ATF3 upregulation in nucleus accumbens decreases emotional reactivity and increases depression-like behavior. Similarly, Turner et al (86) demonstrated that CTGF expression in human amygdala is significantly increased in patients with major depressive disorder compared with healthy subjects. In addition, CTGF administration increases depression-like behavior in outbred rats (86). Only few studies have focused on PLAT association with depression. The results from the present study demonstrated that PLAT, ATF3 and CTGF were co-expressed with SERPINE1, FOS and CCL2. PLAT, ATF3 and CTGF may therefore induce depression through SERPINE1, FOS and CCL2 in patients with OC. However, the underlying mechanisms remain unknown and require further investigation.

Previous studies reported that depression is detrimental to cancer patients' survival (87,88). The results from the present study revealed that upregulation of CCL2, FOS, SERPINE1 and SERPING1 was associated with worse survival. However, Wojnarowicz et al (89) demonstrated that CCL2 protein expression is not correlated with overall or disease-free survival. Conversely, Mahner et al (90) reported that reduced FOS expression is associated with unfavorable PFS and OS in patients with epithelial ovarian carcinoma. However, it has been found that elevated tumor SERPINE1 levels are associated with a poor prognosis and reduced disease-free survival in patients with ovarian carcinoma (91,92). To the best of our knowledge, no study has investigated the association between SERPING1 and survival in patients with OC. However, Peng et al (93) demonstrated that lower SERPING1 mRNA levels predicted worse OS and disease-free survival in patients with prostate cancer compared with healthy controls; however, Mejia et al (94) reported that OS is significantly improved in a mice model of malaria treated with SERPING1 compared with controls. Since it is not clear whether these studies included patients with depression or whether the core genes serve similar functions in different types of cancer, determining how CCL2, FOS, SERPINE1 and SERPING1 could influence patients' survival requires further investigation.

In conclusion, the present study demonstrated that the four core genes CCL2, FOS, SERPINE1 and SERPING1 were upregulated in the HD group, which suggested that they may promote depression and worsen survival in patients with OC through four pathways (‘Ensemble of genes encoding ECM-associated proteins including ECM-affiliated proteins’, ‘ECM regulators and secreted factors’, ‘Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins’ and ‘MAPK signaling pathway and IL-17 signaling pathway’). These findings may provide novel markers and methods for predicting and treating depression in patients with OC; however, the determination of specific mechanisms requires further investigated.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in the published article.

Authors' contributions

YY, YL and WZ designed the study and drafted the manuscript. WW and KW performed the literature search, data extraction and statistical analysis. All authors contributed to the interpretation of the data and the redaction of the manuscript. The final version of the manuscript was reviewed and approved by all authors.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Reid BM, Permuth JB and Sellers TA: Epidemiology of ovarian cancer: A review. Cancer Biol Med. 14:9–32. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in china. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Zhang L, Luo M, Yang H, Zhu S, Cheng X and Qing C: Next-generation sequencing-based genomic profiling analysis reveals novel mutations for clinical diagnosis in chinese primary epithelial ovarian cancer patients. J Ovarian Res. 12:192019. View Article : Google Scholar : PubMed/NCBI

5 

Carlson LE, Angen M, Cullum J, Goodey E, Koopmans J, Lamont L, MacRae JH, Martin M, Pelletier G and Robinson J: High levels of untreated distress and fatigue in cancer patients. Br J Cancer. 90:2297–2304. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Sellick SM and Crooks DL: Depression and cancer: An appraisal of the literature for prevalence, detection, and practice guideline development for psychological interventions. Psychooncology. 8:315–333. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Zabora J, BrintzenhofeSzoc K, Curbow B, Hooker C and Piantadosi S: The prevalence of psychological distress by cancer site. Psychooncology. 10:19–28. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Greimel E, Thiel I, Peintinger F, Cegnar I and Pongratz E: Prospective assessment of quality of life of female cancer patients. Gynecol Oncol. 85:140–147. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Lutgendorf SK, Slavich GM, Degeest K, Goodheart M, Bender D, Thaker PH, Penedo F, Zimmerman B, Lucci J III, Mendez L, et al: Non-cancer life stressors contribute to impaired quality of life in ovarian cancer patients. Gynecol Oncol. 131:667–673. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Watts S, Prescott P, Mason J, McLeod N and Lewith G: Depression and anxiety in ovarian cancer: A systematic review and meta-analysis of prevalence rates. BMJ Open. 5:e0076182015. View Article : Google Scholar : PubMed/NCBI

11 

Huang T, Poole EM, Okereke OI, Kubzansky LD, Eliassen AH, Sood AK, Wang M and Tworoger SS: Depression and risk of epithelial ovarian cancer: Results from two large prospective cohort studies. Gynecol Oncol. 139:481–486. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Mielcarek P, Nowicka-Sauer K and Kozaka J: Anxiety and depression in patients with advanced ovarian cancer: A prospective study. J Psychosom Obstet Gynaecol. 37:57–67. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Wu C, Zhao Y, Liu Y, Yang X, Yan M, Min Y, Pan Z, Qiu S, Xia S, Yu J, et al: Identifying miRNA-mRNA regulation network of major depressive disorder in ovarian cancer patients. Oncol Lett. 16:5375–5382. 2018.PubMed/NCBI

14 

Rahman MR, Islam T, Al-Mamun MA, Zaman T, Karim MR and Moni MA: The influence of depression on ovarian cancer: Discovering molecular pathways that identify novel biomarkers and therapeutic targets. Informatics Med Unlocked. 16:1002072019. View Article : Google Scholar

15 

Watson M, Haviland JS, Greer S, Davidson J and Bliss JM: Influence of psychological response on survival in breast cancer: A population-based cohort study. Lancet. 354:1331–1336. 1999. View Article : Google Scholar : PubMed/NCBI

16 

DiMatteo MR, Lepper HS and Croghan TW: Depression is a risk factor for noncompliance with medical treatment: Meta-analysis of the effects of anxiety and depression on patient adherence. Arch Intern Med. 160:2101–2107. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Yamamoto T, Yanagimoto H, Satoi S, Toyokawa H, Hirooka S, Yamaki S, Yui R, Yamao J, Kim S and Kwon AH: Circulating CD4+CD25+ regulatory T cells in patients with pancreatic cancer. Pancreas. 41:409–415. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lutgendorf SK, DeGeest K, Sung CY, Arevalo JM, Penedo F, Lucci J III, Goodheart M, Lubaroff D, Farley DM, Sood AK and Cole SW: Depression, social support, and beta-adrenergic transcription control in human ovarian cancer. Brain Behav Immun. 23:176–183. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Cole SW, Arevalo JMG, Takahashi R, Sloan EK, Lutgendorf SK, Sood AK, Sheridan JF and Seeman TE: Computational identification of gene-social environment interaction at the human IL6 locus. Proc Natl Acad Sci. 107:5681–5686. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Wang J, Vasaikar S, Shi Z, Greer M and Zhang B: WebGestalt 2017: A more comprehensive, powerful, flexible and interactive gene set enrichment analysis toolkit. Nucleic Acids Res. 45:W130–W137. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Chen J, Bardes EE, Aronow BJ and Jegga AG: ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 37:W305–W311. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res. 41:D991–D995. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Radloff LS: The CES-D scale: A self-report depression scale for research in the general population. Appl Psychol Meas. 1:385–401. 1977. View Article : Google Scholar

24 

Cutroa CE and Rusell DW: The provisions of social relationships and adaptation to stress. Adv Pers Relationships. 1:37–67. 1987.

25 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

26 

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. 43:D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Shannon P, Markiel A, Owen O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Scardoni G, Petterlini M and Laudanna C: Analyzing biological network parameters with centiScaPe. Bioinformatics. 25:2857–2859. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Ogata H, Goto S, Sato K, Fujibuchi W, Bono H and Kanehisa M: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 27:29–34. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Fox R: A view from the web. J R Soc Med. 90:4731997. View Article : Google Scholar

31 

Jia P, Kao CF, Kuo PH and Zhao Z: A comprehensive network and pathway analysis of candidate genes in major depressive disorder. BMC Syst Biol. 5 (Suppl 3):S122011. View Article : Google Scholar : PubMed/NCBI

32 

Hu Y, Pan Z, Hu Y, Zhang L and Wang J: Network and pathway-based analyses of genes associated with parkinson's disease. Mol Neurobiol. 54:4452–4465. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Niwattanakul S, Singthongchai J, Naenudorn E and Wanapu S: Using of jaccard coefficient for keywords similarity. Proc Int Multiconference Eng Comput Sci. 1:13–15. 2013.

34 

Mizuno S, Yamaguchi T, Fukushima A, Matsuyama Y and Ohashi Y: Overlap coefficient for assessing the similarity of pharmacokinetic data between ethnically different populations. Clin Trials. 2:174–181. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Gyorffy B, Lánczky A and Szállási Z: Implementing an online tool for genome-wide validation of survival-associated biomarkers in ovarian-cancer using microarray data from 1287 patients. Endocr Relat Cancer. 19:197–208. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Cancer Genome Atlas Research Network, ; Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: The cancer genome atlas pan-cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Denkert C, Budczies J, Darb-Esfahani S, Györffy B, Sehouli J, Könsgen D, Zeillinger R, Weichert W, Noske A, Buckendahl AC, et al: A prognostic gene expression index in ovarian cancer-validation across different independent data sets. J Pathol. 218:273–280. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Ahmed AA, Mills AD, Ibrahim AE, Temple J, Blenkiron C, Vias M, Massie CE, Iyer NG, McGeoch A, Crawford R, et al: The extracellular matrix protein TGFBI induces microtubule stabilization and sensitizes ovarian cancers to paclitaxel. Cancer Cell. 12:514–527. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Mok SC, Bonome T, Vathipadiekal V, Bell A, Johnson ME, Wong KK, Park DC, Hao K, Yip DK, Donninger H, et al: A gene signature predictive for outcome in advanced ovarian cancer identifies a survival factor: Microfibril-associated glycoprotein 2. Cancer Cell. 16:521–532. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Konstantinopoulos PA, Spentzos D, Karlan BY, Taniguchi T, Fountzilas E, Francoeur N, Levine DA and Cannistra SA: Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol. 28:3555–3561. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Marchion DC, Cottrill HM, Xiong Y, Chen N, Bicaku E, Fulp WJ, Bansal N, Chon HS, Stickles XB, Kamath SG, et al: BAD phosphorylation determines ovarian cancer chemosensitivity and patient survival. Clin Cancer Res. 17:6356–6366. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Gentric G, Kieffer Y, Mieulet V, Goundiam O, Bonneau C, Nemati F, Hurbain I, Raposo G, Popova T, Stern MH, et al: PML-regulated mitochondrial metabolism enhances chemosensitivity in human ovarian cancers. Cell Metab. 29:156–173. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Mateescu B, Batista L, Cardon M, Gruosso T, de Feraudy Y, Mariani O, Nicolas A, Meyniel JP, Cottu P, Sastre-Garau X and Mechta-Grigoriou F: MiR-141 and miR-200a act on ovarian tumorigenesis by controlling oxidative stress response. Nat Med. 17:1627–1635. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Bonome T, Levine DA, Shih J, Randonovich M, Pise-Masison CA, Bogomolniy F, Ozbun L, Brady J, Barrett JC, Boyd J and Birrer MJ: A gene signature predicting for survival in suboptimally debulked patients with ovarian cancer. Cancer Res. 68:5478–5486. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Vathipadiekal V, Wang V, Wei W, Waldron L, Drapkin R, Gillette M, Skates S and Birrer M: Creation of a human secretome: A novel composite library of human secreted proteins: Validation using ovarian cancer gene expression data and a virtual secretome array. Clin Cancer Res. 21:4960–4969. 2015. View Article : Google Scholar : PubMed/NCBI

46 

King ER, Tung CS, Tsang YT, Zu Z, Lok GT, Deavers MT, Malpica A, Wolf JK, Lu KH, Birrer MJ, et al: The anterior gradient homolog 3 (AGR3) gene is associated with differentiation and survival in ovarian cancer. Am J Surg Pathol. 35:904–912. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Ferriss JS, Kim Y, Duska L, Birrer M, Levine DA, Moskaluk C, Theodorescu D and Lee JK: Multi-gene expression predictors of single drug responses to adjuvant chemotherapy in ovarian carcinoma: Predicting platinum resistance. PLoS One. 7:e305502012. View Article : Google Scholar : PubMed/NCBI

48 

Bild AH, Yao G, Chang JT, Wang Q, Potti A, Chasse D, Joshi MB, Harpole D, Lancaster JM, Berchuck A, et al: Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature. 439:353–357. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Koti M, Gooding RJ, Nuin P, Haslehurst A, Crane C, Weberpals J, Childs T, Bryson P, Dharsee M and Evans K: Identification of the IGF1/PI3K/NF κB/ERK gene signalling networks associated with chemotherapy resistance and treatment response in high-grade serous epithelial ovarian cancer. BMC Cancer. 13:5492013. View Article : Google Scholar : PubMed/NCBI

50 

Lisowska KM, Olbryt M, Dudaladava V, Pamuła-Piłat J, Kujawa K, Grzybowska E, Jarząb M, Student S, Rzepecka IK, Jarząb B and Kupryjańczyk J: Gene expression analysis in ovarian cancer-faults and hints from DNA microarray study. Front Oncol. 4:62014. View Article : Google Scholar : PubMed/NCBI

51 

Uehara Y, Oda K, Ikeda Y, Koso T, Tsuji S, Yamamoto S, Asada K, Sone K, Kurikawa R, Makii C, et al: Integrated copy number and expression analysis identifies profiles of whole-arm chromosomal alterations and subgroups with favorable outcome in ovarian clear cell carcinomas. PLoS One. 10:e01280662015. View Article : Google Scholar : PubMed/NCBI

52 

Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, Trivett MK, Etemadmoghadam D, Locandro B, et al: Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res. 14:5198–5208. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. 394–424. 2018.PubMed/NCBI

54 

Chaturvedi SK: Psychiatric oncology: Cancer in mind. Indian J Psychiatry. 54:111–118. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Pasquini M and Biondi M: Depression in cancer patients: A critical review. Clin Pract Epidemiol Ment Health. 3:22007. View Article : Google Scholar : PubMed/NCBI

56 

Prasad SM, Eggener SE, Lipsitz SR, Irwin MR, Ganz PA and Hu JC: Effect of depression on diagnosis, treatment, and mortality of men with clinically localized prostate cancer. J Clin Oncol. 32:2471–2478. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Colombo N, Lorusso D and Scollo P: Impact of recurrence of ovarian cancer on quality of life and outlook for the future. Int J Gynecol Cancer. 27:1134–1140. 2017. View Article : Google Scholar

58 

Leighton SP, Nerurkar L, Krishnadas R, Johnman C, Graham GJ and Cavanagh J: Chemokines in depression in health and in inflammatory illness: A systematic review and meta-analysis. Mol Psychiatry. 23:48–58. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Eyre HA, Air T, Pradhan A, Johnston J, Lavretsky H, Stuart MJ and Baune BT: A meta-analysis of chemokines in major depression. Prog Neuropsychopharmacol Biol Psychiatry. 68:1–8. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Chen K, Pociask DA, McAleer JP, Chan YR, Alcorn JF, Kreindler JL, Keyser MR, Shapiro SD, Houghton AM, Kolls JK and Zheng M: IL-17RA is required for CCL2 expression, macrophage recruitment, and emphysema in response to cigarette smoke. PLoS One. 6:e203332011. View Article : Google Scholar : PubMed/NCBI

61 

Lim SY, Yuzhalin AE, Gordon-Weeks AN and Muschel RJ: Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget. 7:28697–28710. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Yu T, Guo M, Garza J, Rendon S, Sun X, Zhang W and Lu XY: Cognitive and neural correlates of depression-like behaviour in socially defeated mice: An animal model of depression with cognitive dysfunction. Int J Neuropsychopharmacol. 14:303–317. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Kung JC, Chen TC, Shyu BC, Hsiao S and Huang AC: Anxiety- and depressive-like responses and c-fos activity in preproenkephalin knockout mice: Oversensitivity hypothesis of enkephalin deficit-induced posttraumatic stress disorder. J Biomed Sci. 17:292010. View Article : Google Scholar : PubMed/NCBI

64 

Yokoyama K, Hiyama A, Arai F, Nukaga T, Sakai D and Mochida J: C-Fos regulation by the MAPK and PKC pathways in intervertebral disc cells. PLoS One. 8:1–14. 2013. View Article : Google Scholar

65 

Whitmarsh AJ: Regulation of gene transcription by mitogen-activated protein kinase signaling pathways. Biochim Biophys Acta. 1773:1285–1298. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Li JK, Nie L, Zhao YP, Zhang YQ, Wang X, Wang SS, Liu Y, Zhao H and Cheng L: IL-17 mediates inflammatory reactions via p38/c-Fos and JNK/c-Jun activation in an AP-1-dependent manner in human nucleus pulposus cells. J Transl Med. 14:772016. View Article : Google Scholar : PubMed/NCBI

67 

Ma X, Reynolds SL, Baker BJ, Li X, Benveniste EN and Qin H: IL-17 enhancement of the IL-6 signaling cascade in astrocytes. J Immunol. 184:4898–4906. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Hayashi KG, Ushizawa K, Hosoe M and Takahashi T: Differential gene expression of serine protease inhibitors in bovine ovarian follicle: Possible involvement in follicular growth and atresia. Reprod Biol Endocrinol. 9:722011. View Article : Google Scholar : PubMed/NCBI

69 

Lassila M, Fukami K, Jandeleit-Dahm K, Semple T, Carmeliet P, Cooper ME and Kitching AR: Plasminogen activator inhibitor-1 production is pathogenetic in experimental murine diabetic renal disease. Diabetologia. 50:1315–1326. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Yamamoto K, Takeshita K, Shimokawa T, Yi H, Isobe K, Loskutoff DJ and Saito H: Plasminogen activator inhibitor-1 is a major stress-regulated gene: Implications for stress-induced thrombosis in aged individuals. Proc Natl Acad Sci. 99:890–895. 2002. View Article : Google Scholar : PubMed/NCBI

71 

Eskandari F, Mistry S, Martinez PE, Torvik S, Kotila C, Sebring N, Drinkard BE, Levy C, Reynolds JC, Csako G, et al: Younger, premenopausal women with major depressive disorder have more abdominal fat and increased serum levels of prothrombotic factors: Implications for greater cardiovascular risk. Metabolism. 54:918–924. 2005. View Article : Google Scholar : PubMed/NCBI

72 

Lahlou-Laforet K, Alhenc-Gelas M, Pornin M, Bydlowski S, Seigneur E, Benetos A, Kierzin JM, Scarabin PY, Ducimetiere P, Aiach M, et al: Relation of depressive mood to plasminogen activator inhibitor, tissue plasminogen activator, and fibrinogen levels in patients with versus without coronary heart disease. Am J Cardiol. 97:1287–1291. 2006. View Article : Google Scholar : PubMed/NCBI

73 

Tsai SJ, Hong CJ, Liou YJ, Yu YW and Chen TJ: Plasminogen activator inhibitor-1 gene is associated with major depression and antidepressant treatment response. Pharmacogenet Genomics. 18:869–875. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Fang Y, Zhang L, Zeng Z, Lian Y, Jia Y, Zhu H and Xu Y: Promoter polymorphisms of SERPINE1 are associated with the antidepressant response to depression in alzheimer's disease. Neurosci Lett. 516:217–220. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Haslund D, Ryø LB, Seidelin Majidi S, Rose I, Skipper KA, Fryland T, Bohn AB, Koch C, Thomsen MK, Palarasah Y, et al: Dominant-negative SERPING1 variants cause intracellular retention of C1 inhibitor in hereditary angioedema. J Clin Invest. 129:388–405. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Madsen DE, Hansen S, Gram J, Bygum A, Drouet C and Sidelmann JJ: Presence of C1-inhibitor polymers in a subset of patients suffering from hereditary angioedema. PLoS One. 9:e1120512014. View Article : Google Scholar : PubMed/NCBI

77 

Lumry WR, Craig T, Zuraw B, Longhurs H, Baker J, Li HH, Bernstein JA, Anderson J, Riedl MA, Manning ME, et al: Health-related quality of life with subcutaneous C1-inhibitor for prevention of attacks of hereditary angioedema. J Allergy Clin Immunol Pract. 112:1733–1741. 2018. View Article : Google Scholar

78 

Fouche AS, Saunders EF and Craig T: Depression and anxiety in patients with hereditary angioedema. Ann Allergy Asthma Immunol. 112:371–375. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Varga L, Széplaki G, Laki J, Kocsis A, Kristóf K, Gál P, Bajtay Z, Wieslander J, Daha MR, Garred P, et al: Depressed activation of the lectin pathway of complement in hereditary angioedema. Clin Exp Immunol. 153:68–74. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Ditzen C, Tang N, Jastorff AM, Teplytska L, Yassouridis A, Maccarrone G, Uhr M, Bronisch T, Miller CA, Holsboer F and Turck CW: Cerebrospinal fluid biomarkers for major depression confirm relevance of associated pathophysiology. Neuropsychopharmacology. 37:1013–1025. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Liu RM: Oxidative stress, plasminogen activator inhibitor 1, and lung fibrosis. Antioxid Redox Signal. 10:303–319. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Oh CK, Ariue B, Alban RF, Shaw B and Cho SH: PAI-1 promotes extracellular matrix deposition in the airways of a murine Asthma model. Biochem Biophys Res Commun. 294:1155–1160. 2002. View Article : Google Scholar : PubMed/NCBI

83 

Singer M and Jones AM: Bench-to-bedside review: The role of C1-esterase inhibitor in sepsis and other critical illnesses. Crit Care. 15:2032011. View Article : Google Scholar : PubMed/NCBI

84 

Davis AE III, Lu F and Mejia P: C1 inhibitor, a multi-functional serine protease inhibitor. Thromb Haemost. 104:886–893. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Green TA, Alibhai IN, Unterberg S, Neve RL, Ghose S, Tamminga CA and Nestler EJ: Induction of activating transcription factors (ATFs) ATF2, ATF3, and ATF4 in the nucleus accumbens and their regulation of emotional behavior. J Neurosci. 28:2025–2032. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Turner CA, Sharma V, Hagenauer MH, Chaudhury S, O'Connor AM, Hebda-Bauer EK, Thompson RC, Myers RM, Bunney WE and Barchas JD: Connective tissue growth factor is a novel prodepressant. Biol Psychiatry. 84:555–562. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Giese-Davis J, Collie K, Rancourt KM, Neri E, Kraemer HC and Spiegel D: Decrease in depression symptoms is associated with longer survival in patients with metastatic breast cancer: A secondary analysis. J Clin Oncol. 29:413–420. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Raison CL and Miller AH: The evolutionary significance of depression in pathogen host defense (PATHOS-D). Mol Psychiatry. 18:15–37. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Wojnarowicz P, Gambaro K, de Ladurantaye M, Quinn MC, Provencher D, Mes-Masson AM and Tonin PN: Overexpressing the CCL2 chemokine in an epithelial ovarian cancer cell line results in latency of in vivo tumourigenicity. Oncogenesis. 1:e272012. View Article : Google Scholar : PubMed/NCBI

90 

Mahner S, Baasch C, Schwarz J, Hein S, Wölber L, Jänicke F and Milde-Langosch K: C-Fos expression is a molecular predictor of progression and survival in epithelial ovarian carcinoma. Br J Cancer. 99:1269–1275. 2008. View Article : Google Scholar : PubMed/NCBI

91 

Andreasen PA, Kjøller L, Christensen L and Duffy MJ: The urokinase-type plasminogen activator system in cancer metastasis: A review. Int J cancer. 72:1–22. 1997. View Article : Google Scholar : PubMed/NCBI

92 

Klein RM, Bernstein D, Higgins SP, Higgins CE and Higgins PJ: SERPINE1 expression discriminates site-specific metastasis in human melanoma. Exp Dermatol. 21:551–554. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Peng S, Du T, Wu W, Chen X, Lai Y, Zhu D, Wang Q, Ma X, Lin C, Li Z, et al: Decreased expression of serine protease inhibitor family G1 (SERPING1) in prostate cancer can help distinguish high-risk prostate cancer and predicts malignant progression. Urol Oncol. 36:366.e1–366.e9. 2018. View Article : Google Scholar

94 

Mejia P, Diez-Silva M, Kamena F, Lu F, Fernandes SM, Seeberger PH, Davis AE III and Mitchell JR: Human C1-inhibitor suppresses malaria parasite invasion and cytoadhesion via binding to parasite glycosylphosphatidylinositol and host cell receptors. J Infect Dis. 213:80–89. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yi Y, Liu Y, Wu K, Wu W and Zhang W: The core genes involved in the promotion of depression in patients with ovarian cancer. Oncol Lett 18: 5995-6007, 2019
APA
Yi, Y., Liu, Y., Wu, K., Wu, W., & Zhang, W. (2019). The core genes involved in the promotion of depression in patients with ovarian cancer. Oncology Letters, 18, 5995-6007. https://doi.org/10.3892/ol.2019.10934
MLA
Yi, Y., Liu, Y., Wu, K., Wu, W., Zhang, W."The core genes involved in the promotion of depression in patients with ovarian cancer". Oncology Letters 18.6 (2019): 5995-6007.
Chicago
Yi, Y., Liu, Y., Wu, K., Wu, W., Zhang, W."The core genes involved in the promotion of depression in patients with ovarian cancer". Oncology Letters 18, no. 6 (2019): 5995-6007. https://doi.org/10.3892/ol.2019.10934