IMR Press / FBL / Volume 28 / Issue 9 / DOI: 10.31083/j.fbl2809220
Open Access Original Research
NOTCH Pathway Genes in Ovarian Cancer: Clinical Significance and Associations with Immune Cell Infiltration
Show Less
1 The First Clinical Medical College, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
2 Department of Gynecology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
3 Graduate School, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
*Correspondence: zku4ba@163.com (Buze Chen); 446796327@qq.com (Li Jing); 18052268119@189.cn (Xiaoyuan Lu)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(9), 220; https://doi.org/10.31083/j.fbl2809220
Submitted: 19 December 2022 | Revised: 8 May 2023 | Accepted: 12 May 2023 | Published: 24 September 2023
(This article belongs to the Special Issue Diagnostic, Prognostic, and Therapeutic Biomarkers for Ovarian Cancer)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Activation of the NOTCH signaling pathway is associated with tumorigenesis. The aim of this study was to investigate NOTCH pathway gene functions and regulatory mechanisms in ovarian cancer (OC). Methods: We conducted a bioinformatics analysis of publicly available datasets in order to identify potential NOTCH-related mechanisms, associated genes, biological pathways, and their relation to immune function. Results: Significant differential expression of the NOTCH pathway genes DLL1, DLL3, DLL4, HES1, HEY1, JAG1, NOTCH2, NOTCH3, and NOTCH4 was observed between OC samples and normal controls. Low expression of DLL4 and of NOTCH4 in OC patients was associated with International Federation of Gynecology and Obstetrics (FIGO) stage (p <0.001 and p = 0.036, respectively), while high expression of NOTCH3 was associated with race (p = 0.039) and age (p = 0.044). JAG2 and NOTCH1 expression were significantly associated with progression-free interval (PFI) (p = 0.011 and p = 0.039, respectively). DLL1 (Hazard Ratio (HR): 2.096; 95% CI: 1.522–2.886, p < 0.001) and NOTCH1 (HR: 0.711; 95% CI: 0.514–0.983, p = 0.039) expression were independently associated with PFI in multivariate analysis. DLL1, DLL3, JAG1, JAG2, NOTCH3 and NOTCH4 expression could significantly differentiate OC from non-cancer samples. Genes associated with the NOTCH pathway were mainly enriched in five signaling pathways: the NOTCH signaling pathway, breast cancer, endocrine resistance, Th1 and Th2 cell differentiation, and oxidative phosphorylation. The expression of NOTCH pathway genes was significantly associated with immune cell infiltration. Conclusions: NOTCH pathway genes appear to play an important role in the progression of OC by regulating immune cells, endocrine resistance, Th1 and Th2 cell differentiation, and oxidative phosphorylation. JAG2 and NOTCH1 are potential biomarkers and therapeutic targets for the treatment of OC.

Keywords
NOTCH pathway
bioinformatics analysis
ovarian cancer
prognosis
immune infiltration
1. Introduction

Ovarian cancer (OC) has the highest mortality of all gynecologic cancers [1]. The median age of patients diagnosed with OC is approximately 60 years, with most women diagnosed at advanced stages and with metastases [2]. OC is characterized by high levels of recurrence and metastasis, drug resistance, and mortality [3]. Despite advances in surgical techniques and the application of combination chemotherapy, the 5-year survival rate for patients with advanced OC is only 40–45% [4]. Although a great deal of research has been carried out on the pathogenesis of OC, little is known about its regulation and the underlying mechanisms. Molecular and genomic aberrations in OC data has accumulated thanks to the advent of large, multi-cohort genomic mapping libraries like The Cancer Genome Atlas (TCGA) [5, 6]. However, how the data relates to each other, OC disease state, body processes, and other factors has not been well chacterized. This characterization can provide clues as to mechanisms behind OC, which can lead to developing better treatments. Also, this work can lead to development of biomarkers for early detection of OC. The lack of good techniques for early detection contributes significantly to the low survival rate of OC.

The NOTCH signaling pathway is a highly conserved ligand-receptor signaling pathway involved in various aspects of cancer biology, including cancer stemness, angiogenesis, epithelial-mesenchymal transition (EMT), tumor immunity, and drug resistance [7, 8]. The most interesting members of the NOTCH pathway are the NOTCH receptors (NOTCH1, NOTCH2, NOTCH3, NOTCH4), the serrated typical NOTCH ligands Jagged 1 (JAG1) and Jagged 2 (JAG2), the Delta-like typical NOTCH ligands (DLL1, DLL3, DLL4), and the typical downstream genes known as hair and division enhancer 1 (HES1) and hair/division enhancer-associated YRPW patterned protein 1 (HEY1) [8, 9]. There is evidence that NOTCH signaling plays a pleiotropic role in cancer, with dysregulation leading to reduced proliferation and apoptosis of cancer cells [10, 11]. mRNA expression levels for the four types of NOTCH receptor (NOTCH1-4) have different prognostic significance in OC patients [12]. The NOTCH2/NOTCH3/DLL3/MAML1/ADAM17 signaling network has also been associated with OC [13]. Genes that are associated with NOTCH pathway genes can be identified by bioinformatic analysis. Overall, the mechanism and function of the NOTCH pathway in OC remains unclear, and its mechanism of action has still to be clarified.

Cancer results from the co-evolution of malignant cells and the tumor microenvironment (TME) [14]. Bidirectional interactions between the TME and cancer cells regulate its progression, development, metastasis, invasion, and resistance to therapy [15]. Immune infiltration is a critical part of the TME and plays a central role in OC progression [16, 17]. It is important to realize the immune system plays a significant role in the development and progression of OC, and that its dysregulation can result in immune escape and resistance to therapy [18]. Studies of immune cells in the TME of OC have focused on T cells, DCs, MDSCs, macrophages, NK cells, γδ T cells, and B cells [19]. Therefore, identification of the genes that regulate the immune response is essential for understanding how OC cells evade or even suppress the antitumor immune response. To date, activated NOTCH signaling pathways in the TME have been widely reported in various cancer types [8, 14, 20]. However, so far there have been few studies on the correlations between NOTCH members and immune responses in OC, or on the associated mechanisms.

In the present, study we conducted a comprehensive bioinformatics analysis of public datasets to investigate potential NOTCH-related mechanisms, genes, biological pathways, and their relation to immune function. This study looked toidentify valuable candidate biomarkers or targets for the treatment of OC.

2. Materials and Methods
2.1 cBioPortal Analysis

Mutations in NOTCH pathway genes in OC were studied using the cBio Cancer Genomics Portal (http://cbioportal.org) [21, 22, 23]. We performed visualization and analysis queries based on references [23], with the studied proteins being DLL1 [ENSG00000198719], DLL3 [ENSG00000090932], DLL4 [ENSG00000128917], HES1 [ENSG00000114315], HEY1 [ENSG00000164683], JAG1 [ENSG00000101384], JAG2 [ENSG00000184916], NOTCH1 [ENSG00000148400], NOTCH2 [ENSG00000134250], NOTCH3 [ENSG00000074181], and NOTCH4 [ENSG00000204301].

2.2 Dysregulation of NOTCH Pathway Genes in OC

R (version 3.6.3, statistical analysis and visualization, R Core Team, Vienna, Austria) and ggplot2 (version 3.3.3, for visualization, R Core Team, Vienna, Austria) were used [24]. Using the Toil process, UCSC XENA (https://xenabrowser.net/datapages/) RNAseq data was uniformly processed into TPM format for TCGA and GTEx [22, 24, 25, 26, 27, 28]. Data for OC was extracted from TCGA, while GTEx was used for the corresponding normal tissues [24]. RNAseq data were converted into TPM (transcripts per million reads) format by log2 transformation for the comparison of gene expression between samples [24].

2.3 Correlation Heat Map

Correlations between all pairs of genes in the NOTCH pathway were assessed using Pearson’s correlation coefficient [24]. R (version 3.6.3) and ggplot2 (version 3.3.3) were used for the analysis [24].

2.4 Association of NOTCH Pathway Gene Expression with the Clinical Features of TCGA-OC

R (version 3.6.3) and Basic R package were used for this analysis [24]. The grouping condition was the median.

2.5 Survival Analysis

R (version 3.6.3), survminer package (version 0.4.9, for visualization, R Core Team, Vienna, Austria), and survival package (version 3.2-10, R Core Team, Vienna, Austria) were used for the statistical analysis of survival data [24]. The two subgroups studied were 0–50 and 50–100 [24]. Overall survival (OS) and progression-free interval (PFI) [24] were used for prognostic evaluation. Prognostic information for the controls was provided as supplemental data [20].

2.6 Univariate and Multivariate Cox Regression Analysis

R (version 3.6.3) and survival package (version 3.2-10) were used for regression analysis [24]. Cox regression was used as the statistical method [29, 30]. PFI was used for the type of prognosis. Included variables were FIGO stage, primary therapy outcome, race, age, histologic grade, anatomic neoplasm subdivision, venous invasion, lymphatic invasion, tumor residual, tumor status, and 11 NOTCH pathway genes [23]. Prognostic information from the reference was provided as supplemental data [31].

2.7 Forest Plot

R (version 3.6.3) and ggplot2 package (version 3.3.3) were used for Forest plots [24, 30].

2.8 ROC Curve Analysis

R (version 3.6.3), pROC package (version 1.17.0.1, R Core Team, Vienna, Austria) and ggplot2 package [version 3.3.3] were used for analysis [24]. The clinical variables were tumor and normal [24].

2.9 Correlation Analysis for Genes Associated with NOTCH Pathway Genes

R (version 3.6.3) and stat base package (version 3.6.3) were used for this analysis [24].

2.10 Functional Enrichment Analysis of Genes Associated with NOTCH Pathway Genes

R (version 3.6.3), ggplot2 package (version 3.3.3) and clusterProfiler package (version 3.14.3, R Core Team, Vienna, Austria) were used [24].

2.11 Correlations between NOTCH Pathway Gene Expression and Immune Cells

R (version 3.6.3) and GSVA package (version 1.34.0, R Core Team, Vienna, Austria) were used for this analysis [24, 31]. The immune-infiltration algorithm used was ssGSEA (built-in algorithm in the GSVA package) [24]. The immune cell types studied here include aDC (activated DC), B cells, CD8 T cells, cytotoxic cells, DC, eosinophils, iDC (immature DC), macrophages, mast cells, neutrophils, NK CD56_bright cells, NK CD56_dim cells, NK cells, pDC (Plasmacytoid DC), T cells, T helper cells, Tcm (T central memory), Tem (T effector memory), Tfh (T follicular helper), Tgd (T gamma delta), Th1 cells, Th17 cells, Th2 cells, and Tregs [23, 32]. Markers for the 24 immune cell types were obtained from a previous reference [33].

3. Results
3.1 Alterations in the mRNA Expression of NOTCH Pathway Genes in OC

The online tool cBioPortal was used to examine NOTCH pathway gene alterations in OC. As shown in Fig. 1, the frequency of genetic alterations in OC was: DLL1, 3%; DLL3, 10%; DLL4, 2.8%; HES1, 19%; HEY1, 5%; JAG1, 4%; JAG2, 5%; NOTCH1, 4%; NOTCH2, 8%, NOTCH3, 14%; and NOTCH4, 5%. As shown in Fig. 2, the frequency of genetic alterations in NOTCH pathway genes in OC patients from TCGA, Firehose Legacy were: mutation, 1.2%; amplification, 52.05%; deep deletion, 6.85%; and multiple alterations, 1.71%. For TCGA, Pan-Cancer Atlas: mutation, 3.25%; structural variant, 0.34%; amplification, 37.84%; deep deletion, 5.48%; and multiple alterations 2.91%. For TCGA, Nature 2011: mutation, 1.23%; amplification, 33.74%; deep deletion, 3.68%; and multiple alterations, 1.43%. In general, amplifications accounted for most of the genetic alterations detected in OC.

Fig. 1.

Genetic alterations of NOTCH pathway genes in ovarian cancer (OC) as observed using cBioPortal (RNA Seq V2 RSEM).

Fig. 2.

The distribution of genetic alterations in NOTCH pathway genes in OC cases based on the cancer type summary in cBioPortal.

A total of 427 OC tumors and 88 samples of normal ovarian tissue were used to assess NOTCH pathway gene expression (Fig. 3). The expression levels of DLL1, DLL4, HEY1, JAG1 and NOTCH4 were all significantly lower in OC samples than in normal ovarian tissues (1.064 ± 0.832 vs. 1.991 ± 0.827; 1.688 ± 0.631 vs. 2.206 ± 0.878; 1.33 ± 0.876 vs. 1.51 ± 0.632; 3.339 ± 1.027 vs. 4.916 ± 0.835; and 2.113 ± 0.568 vs. 3.514 ± 0.689, respectively; all p < 0.001). In contrast, the expression levels of DLL3, HES1, JAG2, NOTCH1 and NOTCH3 were all significantly higher in OC tumor samples (1.075 ± 0.884 vs. 0.25 ± 0.246; 4.9 ± 1.119 vs. 4.383 ± 1.168; 3.121 ± 0.929 vs. 2.284 ± 0.731; 2.82 ± 1.063 vs. 2.487 ± 0.676; and 5.086 ± 1.253 vs. 3.933 ± 1.353, respectively; all p < 0.001 except NOTCH1, p = 0.002). No significant difference was seen in the level of NOTCH2 expression between OC tumor tissues and healthy ovarian tissues.

Fig. 3.

TCGA mRNA levels for the NOTCH pathway genes in OC tissue and in unpaired normal ovarian tissue. ns, not significant (p 0.05); **, p < 0.01; ***, p < 0.001.

Pearson correlation analysis was used to test for relationships between genes involved in the NOTCH pathway. A significant negative correlation was found between the expression of DLL3 and NOTCH2, while the expression levels of most other NOTCH pathway genes showed a significant positive correlation with each other (Fig. 4).

Fig. 4.

Correlations between the expression level of genes in the NOTCH pathway.

3.2 Relationship between NOTCH Pathway Gene Expression and the Clinical Characteristics of TCGA OC Patients

The clinical characteristics and gene expression data for 379 OC tumor samples were downloaded from the TCGA database (Supplementary Table 1). DLL4 and NOTCH4 expression were associated with FIGO stage (p < 0.001 and p = 0.036, respectively), while NOTCH3 expression was associated with race (p = 0.039) and age (p = 0.044). The expression levels of other NOTCH pathway genes (DLL1, DLL3, HES1, HEY1, JAG1, JAG2, NOTCH1, and NOTCH2) did not correlate significantly with the clinical data of OC patients.

3.3 Prognostic Value of NOTCH Pathway Genes in OC

The expression level of 11 NOTCH pathway genes (DLL1, DLL3, DLL4, HES1, HEY1, JAG1, JAG2, NOTCH1, NOTCH2, NOTCH3, and NOTCH4) showed no significant correlation with the overall survival of OC patients (Fig. 5). The expression of JAG2 (p = 0.011) and NOTCH1 (p = 0.039) were found to be significantly associated with the PFI of OC patients, whereas the remaining 9 genes (DLL1, DLL3, DLL4, HES1, HEY1, JAG1, NOTCH2, NOTCH3 and NOTCH4) showed no significant correlation with the PFI (Fig. 6). Higher expression of JAG2 and NOTCH1 were correlated with significantly better PFI.

Fig. 5.

Overall survival (OS) analysis for NOTCH pathway gene expression in OC patients using Kaplan Meir plot web tool. (A) DLL1, (B) DLL3, (C) DLL4, (D) HES1, (E) HEY1, (F) JAG1, (G) JAG2, (H) NOTCH1, (I) NOTCH2, (J) NOTCH3, and (K) NOTCH4.

Fig. 6.

Progression-free interval (PFI) analysis for NOTCH pathway gene expression in OC patients using Kaplan Meir plot web tool. (A) DLL1, (B) DLL3, (C) DLL4, (D) HES1, (E) HEY1, (F) JAG1, (G) JAG2, (H) NOTCH1, (I) NOTCH2, (J) NOTCH3, and (K) NOTCH4.

3.4 Univariate and Multivariate Cox Regression Analysis

Univariate Cox regression analysis (Fig. 7A) revealed that primary therapy outcome (HR: 0.401; 95% CI: 0.304–0.528, p < 0.001), JAG2 expression (HR: 0.735; 95% CI: 0.580–0.932, p = 0.011), and NOTCH1 expression (HR: 0.779; 95% CI: 0.614–0.987, p = 0.039) were predictors of good PFI in OC patients. In contrast, tumor residual (HR: 1.695; 95% CI: 1.219–2.358, p = 0.002) and tumor status (HR: 10.045; 95% CI: 5.758–17.526, p < 0.001) were predictors of worse PFI in OC patients. Multivariate analysis revealed that primary therapy outcome (HR: 0.613; 95% CI: 0.451–0.832, p = 0.002), tumor status (HR: 14.164; 95% CI: 6.909–29.039, p < 0.001), DLL1 expression (HR: 2.096; 95% CI: 1.522–2.886, p < 0.001), and NOTCH1 expression (HR: 0.711; 95% CI: 0.514–0.983, p = 0.039) were independent predictors of PFI.

Fig. 7.

Forest plots showing the results of Cox regression analysis for NOTCH pathway genes and other clinical features as predictors of progression free interval (PFI) in OC patients. (A) Univariate regression analysis. (B) Multivariate regression analysis.

3.5 Diagnostic Value of NOTCH Pathway Gene Expression in OC

As shown in Fig. 8, the expression of several NOTCH pathway genes showed some tendency for differentiating normal and OC tumor tissues (DLL1: AUC = 0.823, CI = 0.785–0.861; DLL3: AUC = 0.839, CI = 0.801–0.876; JAG1: AUC = 0.883, CI = 0.849–0.916; JAG2: AUC = 0.769, CI = 0.718–0.820; NOTCH3: AUC = 0.739, CI = 0.679–0.799). Other genes showed low tendency (DLL4: AUC = 0.668, CI = 0.599–0.737; HES1: AUC = 0.642, CI = 0.576–0.709; HEY1: AUC = 0.621, CI = 0.560–0.683; NOTCH1: AUC = 0.603, CI = 0.549–0.657; NOTCH2: AUC = 0.532, CI = 0.482–0.583), while high accuracy was shown by NOTCH4 (AUC = 0.948, CI = 0.928–0.968).

Fig. 8.

ROC curves for NOTCH pathway genes in OC and normal ovarian tissues. The area under the curve (AUC) ranged from 0.5 to 1. When the AUC is closer to 1, diagnosis is more accurate for the differentiation of OC. An AUC between 0.5 and 0.7 has low accuracy, an AUC between 0.7 and 0.9 has some accuracy, while an AUC above 0.9 has high accuracy.

3.6 Functions Associated with NOTCH Pathway Genes

The top five negatively associated genes with each NOTCH pathway gene are shown in Fig. 9, together with the top 5 positively associated genes. The genes most positively associated with DLL1 were ADAMTS7, DENND2A, ZNF697, RHOBTB3 and DCHS1, while those most negatively associated with this gene were RTP4, EVA1C, EIF4EBP3, PDZK1IP1, and PLAAT4. The genes most positively associated with DLL3 were SELENOV, TIMM50, ECH1, RYR1 and MRPS12, while those most negatively associated were VNN1, MVP, KCTD12, SERPINB1, and TMEM87A. The genes most positively associated with DLL4 were NOTCH4, CDH5, VWF, PCDH12 and ROBO4, while those most negatively associated were RPL39, HIGD2A, ATP5MD, RPL36A, and RPS27. The genes most positively associated with HES1 were SMOX, DACT2, JAG1, ITM2C, and PDE9A, while those most negatively associated were HPSE, LTC4S, NPR1, CYP4B1, and EVA1C. The genes most positively associated with HEY1 were PGF, PEAR1, TSPAN11, HSPA12B, and SIX2, while those most negatively associated were KLK7, PDZK1IP1, C19orf33, FOLR1, and MSLN. The genes most positively associated with JAG1 were NOTCH1, BTBD3, NOTCH3, ATRN, and CDS2, while those most negatively associated were COX5B, ATP5MC2, LAMTOR4, RPL32, NDUFA1. The genes most positively associated with JAG2 were CEP170B, BRF1, AKT1, PACS2, and PLCB3, while those most negatively associated were OST4, COPS9, BLOC1S1, COX7C, and PFDN5. The genes most positively associated with NOTCH1 were SEC16A, CAMSAP1, PLXNA1, RXRA and PRRC2B, while those most negatively associated were ATP5MF, UQCRQ, NDUFA1, FAU and COX5B. The genes most positively associated with NOTCH2 were CTDSP2, HIPK1, GIT2, ATXN1L and BAZ2A, while those most negatively associated were RNF181, ATP5MD, COX5B, ATP5F1E and RPS21. The genes most positively associated with NOTCH3 were BRD4, WIZ, MYO9B, TET3 and CEP250, while those most negatively associated were VAMP8, UQCR11, PLAAT4, COX7B, and NDUFA1. The genes most positively associated with NOTCH4 were CDH5, ROBO4, PCDH12, DIPK2B, and DLL4, while those most negatively associated were RPL12, RPL36A, RPL31, RPS21 and RPS27.

Fig. 9.

Heatmap showing the top 10 genes that were correlated with NOTCH pathway genes. (A) DLL1, (B) DLL3, (C) DLL4, (D) HES1, (E) HEY1, (F) JAG1, (G) JAG2, (H) NOTCH1, (I) NOTCH2, (J) NOTCH3, and (K) NOTCH4.

GO and KEGG enrichment analyses were conducted on the NOTCH pathway and related genes (102 genes in total) (Supplementary Table 2). As shown in Fig. 10 and Supplementary Table 3, the top five biological processes identified were the regulation of NOTCH signaling pathway, NOTCH signaling pathway, negative regulation of NOTCH signaling pathway, NOTCH signaling involved in heart development, and positive regulation of NOTCH signaling pathway. The top five cytological components were the mitochondrial inner membrane, polysomal ribosome, cytosolic ribosome, mitochondrial proton-transporting ATP synthase complex, and cytosolic part. The top five molecular functions were NOTCH binding, structural constituent of ribosome, electron transfer activity, cytochrome-c oxidase activity, and heme-copper terminal oxidase activity. As shown in Fig. 11 and Supplementary Table 3, the top five pathways identified were the NOTCH signaling pathway, breast cancer, endocrine resistance, Th1 and Th2 cell differentiation, and oxidative phosphorylation.

Fig. 10.

GO analysis of genes involved in the NOTCH pathway.

Fig. 11.

KEGG analysis of genes associated with NOTCH pathway genes.

3.7 NOTCH Pathway Gene Expression in OC and Correlation with Immune Cells

As shown in Fig. 12, NOTCH pathway gene expression was correlated with tumor-infiltrating immune cells (TIIC) in OC. DLL1 gene expression correlated positively with eosinophils, iDC, macrophages, mast cells, NK cells, Tcm, Tem, TFH and Tgd, and negatively with aDC, cytotoxic cells, T cells, Th1 cells, and Th17 cells. DLL3 expression correlated positively with Th2 cells, and negatively with CD8 T cells, cytotoxic cells, DC, iDC, macrophages, neutrophils, NK CD56bright cells, NK CD56dim cells, pDC, T cells, Tcm, Tem, Th1 cells and Th17 cells. DLL4 expression correlated positively with eosinophils, iDC, macrophages, mast cells, neutrophils, NK cells, pDC, T helper cells, Tcm, Tem, TFH and Tgd, and negatively with cytotoxic cells. HES1 expression correlated positively with eosinophils, NK cells and Th2 cells, and negatively with aDC, B cells, cytotoxic cells, DC, neutrophils, T cells, Tem, Th1 cells, Th17 cells and Tregs. HEY1 expression correlated positively with eosinophils, iDC, macrophages, mast cells, neutrophils, NK cells, T helper cells, Tem, TFH, Tgd and Th2 cells, and negatively with cytotoxic cells, NK CD56bright cells and Th17 cells. JAG1 expression correlated positively with eosinophils, iDC, NK cells, T helper cells, Tcm, Tem, TFH and Th2 cells, and negatively with cytotoxic cells. JAG2 expression correlated positively with eosinophils, NK cells, T helper cells, Tcm and Tem. NOTCH1 expression correlated positively with DC, eosinophils, iDC, macrophages, mast cells, neutrophils, NK CD56dim cells, NK cells, T helper cells, Tcm, Tem, TFH and Th2 cells, and negatively with cytotoxic cells. NOTCH2 expression correlated positively with DC, eosinophils, iDC, macrophages, mast cells, neutrophils, NK cells, T helper cells, Tcm, Tem, TFH, Tgd and Th17 cells. NOTCH3 expression correlated positively with eosinophils, NK cells, Tcm and Tem, and negatively with aDC, CD8 T cells, cytotoxic cells, pDC, T cells, Th1 cells, Th17 cells, and T regs. NOTCH4 gene expression correlated positively with eosinophils, iDC, macrophages, mast cells, neutrophils, NK cells, pDC, T helper cells, Tcm, Tem, TFH and Tgd.

Fig. 12.

Analysis of the correlation between expression of each NOTCH family gene and 24 types of TIICs in OC. (A) DLL1, (B) DLL3, (C) DLL4, (D) HES1, (E) HEY1, (F) JAG1, (G) JAG2, (H) NOTCH1, (I) NOTCH2, (J) NOTCH3, and (K) NOTCH4. Results are shown in the form of lollipop plots. The darker the color, the smaller the p-value and hence greater statistical significance. Correlations are represented by bubble sizes, with larger bubbles indicating stronger correlation.

4. Discussion

OC patients with NOTCH3 alterations, including upregulation, experience poor survival, while the targeting of NOTCH3 inhibits cell growth and induces apoptosis [34]. Associations have been reported between the expression of OCT4, NOTCH1 and DLL4 with OC differentiation, lymph node metastasis, clinical stage, and prognosis [35]. High expression of NOTCH2 and NOTCH3 mRNA in OC patients has been significantly associated with PFS, while high expression of NOTCH4 mRNA has been significantly associated with OS [12]. Five genes (NOTCH2, NOTCH3, DLL1, DLL3, and DLL4) have also been associated with poorer survival in OC [35]. In the present study, significant differential expression between OC samples and controls was observed for the NOTCH pathway genes DLL1, DLL3, DLL4, HES1, HEY1, JAG1, NOTCH2, NOTCH3, and NOTCH4. DLL4 expression was associated with FIGO stage (p < 0.001), while high NOTCH3 expression was associated with race (p = 0.039) and age (p = 0.044). Low expression of NOTCH4 was also associated with FIGO stage (p = 0.036). There were significant differences in PFI between patients with low and high expression of JAG1 (p = 0.011) and NOTCH1 (p = 0.039). Multivariate analysis revealed that DLL1 (HR: 2.096; 95% CI: 1.522–2.886, p < 0.001) and NOTCH1 (HR: 0.711; 95% CI: 0.514–0.983, p = 0.039) expression were independent predictors of PFI. Moreover, the expression of DLL1, DLL3, JAG1, JAG2, NOTCH3 and NOTCH4 could significantly differentiate between cancerous and non-cancerous tissue samples.

Estrogen promotes the differentiation of ovarian multi-ciliated cells (MCC) by reducing DLL1 expression via the estrogen receptor β [36]. Sequential combination of cisplatin/eugenol targets the NOTCH-Hes1 pathway and eliminates drug-resistant cancer stem cells [37]. OC progression and resistance have been linked to JAG1, an oncogene, and hence targeting the GATA1/JAG1/NOTCH pathway might represent a new treatment strategy [38]. JAG1 inhibits OC cell growth and may act by suppressing the NOTCH1 signaling pathway [39]. JAG1/NOTCH3 interactions constitute a juxtaposed secretory loop that promotes the proliferation and dissemination of OC cells in the peritoneal cavity [40]. Thus, BMP9-NOTCH1 signaling could also be a new therapeutic target axis for OC treatment [41]. MiR-34b inhibits proliferation and epithelial-mesenchymal transition in OC by targeting NOTCH2 [42]. Activation of NOTCH3-mediated signaling by IL-8 secreted from cancer-associated fibroblasts and cancer cells promotes stem cell sexuality in OC [43]. In the present study, genes associated with the NOTCH pathway were mainly enriched in five signaling pathways. These were the NOTCH signaling pathway, breast cancer, endocrine resistance, Th1 and Th2 cell differentiation, and oxidative phosphorylation. In conclusion, these findings suggest the NOTCH pathway could regulate OC progression by modulating endocrine resistance, Th1 and Th2 cell differentiation, and oxidative phosphorylation. Therefore, targeting of the cross-talk network rather than a single pathway may be a more effective treatment strategy.

The tumor infiltration of immune cells and the immune evasion by cancer cells play key roles in tumor progression [9]. JAG2+ TANs are closely associated with the IL-8-driven immune evasion microenvironment, thus providing a potential therapeutic target for enhancing the immunity against OC [44]. Glycolytic targeting of microRNA and EZH2 in OC has been reported to lead to effector T cell dysfunction [45]. In the present study, expression of the DLL1, HES1 and NOTCH3 genes was negatively associated with T cell infiltration, whereas expression of the DLL4, HEY1, JAG1, JAG2, NOTCH1, NOTCH2 and NOTCH4 genes was positively associated with T cells. In contrast, DLL3 expression was positively correlated with Th2 cells and negatively correlated with other types of T cells. The use of adaptive T cells, such as chimeric antigen receptor T cell therapy, may therefore be a promising new paradigm for the treatment of OC. A better understanding of NOTCH signaling could improve therapeutic approaches.

In summary, we evaluated the expression levels, mutations, and immune correlations of the NOTCH pathway in OC in order to identify potential biomarkers and targets. In addition to improving clinical decision making, the results contribute to a deeper understanding of the complex involvement of the NOTCH pathway in OC. Future studies should investigate the role of NOTCH signaling in OC in vitro and in vivo, as none were performed in the present study.

5. Conclusions

Activation of NOTCH plays an important role in mediating the development and progression of OC through multiple pathways, including the regulation of immune cells, endocrine resistance, Th1 and Th2 cell differentiation, and oxidative phosphorylation. Downregulation of JAG2 and NOTCH1 expression were associated with significantly worse PFI in OC patients. JAG2 and NOTCH1 may be potentially useful biomarkers for the treatment of OC, or as therapeutic targets.

Availability of Data and Materials

The datasets for this study can be found in TCGA-OC (https://portal.gdc.cancer.gov/). All data generated or analyzed during this study are included in this article.

Author Contributions

BC, LJ, and XYL designed the research study. KJ, HW, and LM analyzed the data. XML and QC collected the data. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA: A Cancer Journal for Clinicians. 2020; 70: 7–30.
[2]
Wu A, Zhang S, Liu J, Huang Y, Deng W, Shu G, et al. Integrated Analysis of Prognostic and Immune Associated Integrin Family in Ovarian Cancer. Frontiers in Genetics. 2020; 11: 705.
[3]
Webb PM, Jordan SJ. Epidemiology of epithelial ovarian cancer. Best Practice & Research. Clinical Obstetrics & Gynaecology. 2017; 41: 3–14.
[4]
Henderson JT, Webber EM, Sawaya GF. Screening for Ovarian Cancer: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2018; 319: 595–606.
[5]
Li H, Li M, Tang C, Xu L. Screening and prognostic value of potential biomarkers for ovarian cancer. Annals of Translational Medicine. 2021; 9: 1007.
[6]
Lu X, Li G, Liu S, Wang H, Zhang Z, Chen B. Bioinformatics Analysis of KIF1A Expression and Gene Regulation Network in Ovarian Carcinoma. International Journal of General Medicine. 2021; 14: 3707–3717.
[7]
Fasoulakis Z, Daskalakis G, Theodora M, Antsaklis P, Sindos M, Diakosavvas M, et al. The Relevance of NOTCH Signaling in Cancer Progression. Advances in Experimental Medicine and Biology. 2021; 1287: 169–181.
[8]
Zhang C, Berndt-Paetz M, Neuhaus J. A Comprehensive Bioinformatics Analysis of NOTCH Pathways in Bladder Cancer. Cancers. 2021; 13: 3089.
[9]
Xue D, Li D, Dou C, Li J. A Comprehensive Bioinformatic Analysis of NOTCH Pathway Involvement in Stomach Adenocarcinoma. Disease Markers. 2021; 2021: 4739868.
[10]
Roy M, Pear WS, Aster JC. The multifaceted role of NOTCH in cancer. Current Opinion in Genetics & Development. 2007; 17: 52–59.
[11]
Pinnix CC, Herlyn M. The many faces of NOTCH signaling in skin-derived cells. Pigment Cell Research. 2007; 20: 458–465.
[12]
Chen C, Wang X, Huang S, Wang L, Han L, Yu S. Prognostic roles of NOTCH receptor mRNA expression in human ovarian cancer. Oncotarget. 2017; 8: 32731–32740.
[13]
Jia D, Underwood J, Xu Q, Xie Q. NOTCH2/NOTCH3/DLL3/MAML1/ADAM17 signaling network is associated with ovarian cancer. Oncology Letters. 2019; 17: 4914–4920.
[14]
Meurette O, Mehlen P. NOTCH Signaling in the Tumor Microenvironment. Cancer Cell. 2018; 34: 536–548.
[15]
Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012; 21: 309–322.
[16]
Nowak M, Klink M. The Role of Tumor-Associated Macrophages in the Progression and Chemoresistance of Ovarian Cancer. Cells. 2020; 9: 1299.
[17]
Yang L, Wang S, Zhang Q, Pan Y, Lv Y, Chen X, et al. Clinical significance of the immune microenvironment in ovarian cancer patients. Molecular Omics. 2018; 14: 341–351.
[18]
Farolfi A, Gurioli G, Fugazzola P, Burgio SL, Casanova C, Ravaglia G, et al. Immune System and DNA Repair Defects in Ovarian Cancer: Implications for Locoregional Approaches. International Journal of Molecular Sciences. 2019; 20: 2569.
[19]
Lei X, Lei Y, Li JK, Du WX, Li RG, Yang J, et al. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Letters. 2020; 470: 126–133.
[20]
Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell. 2018; 173: 400–416.e11.
[21]
Zhang C, Cui Y, Wang G, Zhao W, Zhao H, Huang X, et al. Comprehensive Analysis of the Expression and Prognosis for E2Fs in Human Clear Cell Renal Cell Carcinoma. Journal of Healthcare Engineering. 2021; 2021: 5790416.
[22]
Hu Y, Wang M, Wang K, Gao J, Tong J, Zhao Z, et al. A potential role for metastasis-associated in colon cancer 1 (MACC1) as a pan-cancer prognostic and immunological biomarker. Mathematical Biosciences and Engineering: MBE. 2021; 18: 8331–8353.
[23]
Chen B, Gao C, Wang H, Sun J, Han Z. Molecular Analysis of Prognosis and Immune Infiltration of Ovarian Cancer Based on Homeobox D Genes. Computational and Mathematical Methods in Medicine. 2022; 2022: 3268386.
[24]
Chen Y, Li D, Wang D, Peng H. Comprehensive analysis of Distal-Less homeobox family gene expression in colon cancer. Research Square. 2022. (preprint)
[25]
Vivian J, Rao AA, Nothaft FA, Ketchum C, Armstrong J, Novak A, et al. Toil enables reproducible, open source, big biomedical data analyses. Nature Biotechnology. 2017; 35: 314–316.
[26]
Yi W, Shen H, Sun D, Xu Y, Feng Y, Li D, et al. Low Expression of Long Noncoding RNA SLC26A4 Antisense RNA 1 Is an Independent Prognostic Biomarker and Correlate of Immune Infiltrates in Breast Cancer. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research. 2021; 27: e934522.
[27]
Yang D, Liu M, Jiang J, Luo Y, Wang Y, Chen H, et al. Comprehensive Analysis of DMRT3 as a Potential Biomarker Associated with the Immune Infiltration in a Pan-Cancer Analysis and Validation in Lung Adenocarcinoma. Cancers. 2022; 14: 6220.
[28]
Liang W, Lu Y, Pan X, Zeng Y, Zheng W, Li Y, et al. Decreased Expression of a Novel lncRNA FAM181A-AS1 is Associated with Poor Prognosis and Immune Infiltration in Lung Adenocarcinoma. Pharmacogenomics and Personalized Medicine. 2022; 15: 985–998.
[29]
Li Y, Li JQ, Jiang HP, Li X. The Upregulation of PLXDC2 Correlates with Immune Microenvironment Characteristics and Predicts Prognosis in Gastric Cancer. Disease Markers. 2021; 2021: 5669635.
[30]
Han QL, Cui Z, Wang Q, Pang F, Li D, Wang D. Upregulation of OTX2-AS1 is Associated with Immune Infiltration and Predicts Prognosis of Gastric Cancer. Technology in Cancer Research & Treatment. 2023; 22: 15330338231154091.
[31]
Hänzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013; 14: 7.
[32]
Zhang C, Zhang W, Cui H, Zhang B, Miao P, Yang Q, et al. Role of Hub Genes in the Occurrence and Development of Testicular Cancer Based on Bioinformatics. International Journal of General Medicine. 2022; 15: 645–660.
[33]
Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013; 39: 782–795.
[34]
Hu W, Liu T, Ivan C, Sun Y, Huang J, Mangala LS, et al. NOTCH3 pathway alterations in ovarian cancer. Cancer Research. 2014; 74: 3282–3293.
[35]
Yu L, Jiao YJ, Zhou L, Song WQ, Wu SW, Wang DN. Expressions of OCT4, NOTCH1 and DLL4 and their clinical implications in epithelial ovarian cancer. Nan Fang Yi Ke Da Xue Xue Bao. 2016; 37: 444–450. (In Chinese)
[36]
Zhu M, Iwano T, Takeda S. Estrogen and EGFR Pathways Regulate NOTCH Signaling in Opposing Directions for Multi-Ciliogenesis in the Fallopian Tube. Cells. 2019; 8: 933.
[37]
Islam SS, Aboussekhra A. Sequential combination of cisplatin with eugenol targets ovarian cancer stem cells through the NOTCH-Hes1 signalling pathway. Journal of Experimental & Clinical Cancer Research: CR. 2019; 38: 382.
[38]
Liu Z, Zhu Y, Li F, Xie Y. GATA1-regulated JAG1 promotes ovarian cancer progression by activating NOTCH signal pathway. Protoplasma. 2020; 257: 901–910.
[39]
Liu GY, Gao ZH, Li L, Song TT, Sheng XG. Expression of Jagged1 mRNA in human epithelial ovarian carcinoma tissues and effect of RNA interference of Jagged1 on growth of xenograft in nude mice. Zhonghua Fu Chan Ke Za Zhi. 2016; 51: 448–453. (In Chinese)
[40]
Choi JH, Park JT, Davidson B, Morin PJ, Shih IM, Wang TL. Jagged-1 and NOTCH3 juxtacrine loop regulates ovarian tumor growth and adhesion. Cancer Research. 2008; 68: 5716–5723.
[41]
Yang L, Bai Y, Zhang C, Du J, Cheng Y, Wang Q, et al. Overexpression of BMP9 promotes ovarian cancer progression via NOTCH1 signaling. Neoplasma. 2021; 68: 1190–1200.
[42]
Lu S, Liu W, Shi H, Zhou H. Exosomal miR-34b inhibits proliferation and the epithelial-mesenchymal transition by targeting NOTCH2 in ovarian cancer. Oncology Letters. 2020; 20: 2721–2728.
[43]
Ji Z, Tian W, Gao W, Zang R, Wang H, Yang G. Cancer-Associated Fibroblast-Derived Interleukin-8 Promotes Ovarian Cancer Cell Stemness and Malignancy Through the NOTCH3-Mediated Signaling. Frontiers in Cell and Developmental Biology. 2021; 9: 684505.
[44]
Yang M, Zhang G, Wang Y, He M, Xu Q, Lu J, et al. Tumour-associated neutrophils orchestrate intratumoural IL-8-driven immune evasion through Jagged2 activation in ovarian cancer. British Journal of Cancer. 2020; 123: 1404–1416.
[45]
Zhao E, Maj T, Kryczek I, Li W, Wu K, Zhao L, et al. Cancer mediates effector T cell dysfunction by targeting microRNAs and EZH2 via glycolysis restriction. Nature Immunology. 2016; 17: 95–103.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top