Open Access

The diagnostic or prognostic values of FADD in cancers based on pan‑cancer analysis

  • Authors:
    • Chenyu Wang
    • Xianglai Jiang
    • Qiqi Zhao
    • Zhiyuan Xie
    • Hui Cai
  • View Affiliations

  • Published online on: September 11, 2023     https://doi.org/10.3892/br.2023.1659
  • Article Number: 77
  • Copyright: © Wang 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

Previous studies have determined that aberrant expression of the fas‑associated death domain (FADD) contributes to the development of cancer. However, no pan‑cancer analysis has been reported to explore the relationship between FADD and various cancers. Multiple databases were screened to identify cancer datasets for the present study and to validate the expression of FADD in various tumors. The association of FADD alteration with cancer prognosis, clinical features and tumor immunity was also evaluated. Reverse transcription‑quantitative PCR (RT‑qPCR) was utilized to confirm the expression of FADD in breast, colon, liver and gastric cancer cells. Analysis of Gene Expression Omnibus database and The Cancer Genome Atlas database indicated that FADD was highly expressed in breast invasive carcinoma (BRCA), cervical squamous cell carcinoma and endocervical adenocarcinoma, cholangiocarcinoma, colon adenocarcinoma (COAD), esophageal carcinoma (ESCA), kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma (LIHC), lung adenocarcinoma (LUAD) and prostate adenocarcinoma, whereas RT‑qPCR results revealed that FADD was highly expressed in breast cancer and colon cancer. Further analyses demonstrated that FADD expression was significantly altered in ESCA, head and neck squamous cell carcinoma (HNSC), lung squamous cell carcinoma and BRCA. FADD expression was observed to be a risk factor of the overall survival in patients with HNSC, LIHC and LUAD as demonstrated by Kaplan‑Meier and Cox regression analyses. The results of the present study demonstrated that FADD is highly expressed in numerous malignancies and can be utilized as a biomarker for the diagnosis of BRCA, COAD, LIHC and stomach adenocarcinoma. Moreover, FADD expression is a predictive risk factor for the development of HNSC, LIHC and LUAD and can potentially be used as a prognostic marker for these cancers.

Introduction

Fas-associated death domain (FADD) is a ubiquitous adaptor protein (1). The human FADD gene consists of two exons and one intron and has been mapped to chromosome 11q13.3, a region strongly associated with breast invasive carcinoma (BRCA), lung cancer and esophageal carcinoma (ESCA) (2). As an important receptor protein in the tumor necrosis factor receptor family-mediated apoptosis pathway, FADD modulates its binding to death receptors of the tumor necrosis factor receptor family to transmit apoptosis initiation signals (3-5). In addition, FADD is involved in the regulation of cell proliferation, gene expression and immunity (1,5-8). As a universal adaptor molecule, abnormal expression of FADD protein is associated with the occurrence and development of tumors in both mature and embryonic tissues.

In the last few years, major breakthroughs have been made in the treatment of cancer, including immunotherapy, which has achieved remarkable results in clinical practice (9,10). As the most important defense system of the human organism, the immune system does not only eliminate pathogenic microorganisms, but also destroys abnormal cancer cells, thus actively inhibits tumor growth (11). However, the composition of tumors and their related tumor microenvironment (TME) is relatively complex, requiring precise immune responses (11,12). Therefore, cancer immunotherapy can only achieve favorable results in specific cancer types and patients (13,14). Research to find potential targets for cancer immunotherapy and predict its efficacy is critical to achieve specificity in cancer treatment. Previous studies have demonstrated that FADD is involved in and regulates signaling complexes, including necrosomes, endosomes and inflammasomes (1,15,16). Thus, FADD plays an indispensable role in innate immunity, inflammation and cancer development (1). However, the role of FADD in tumorigenesis is not fully understood, and whether it can be used as a prognostic biomarker as well as its potential value for clinical treatment require to be further explored. In the present study, the differential expression, gene alteration, prognostic value, tumor progression and promoter methylation level of FADD in pan-cancer extent were evaluated based on The Cancer Genome Atlas (TCGA) dataset. Subsequently, the expression level of FADD in related cell lines and databases as well as its relationship with immune cell infiltration, immune checkpoint, tumor mutation burden (TMB) and microsatellite instability (MSI) were analyzed.

Materials and methods

Cell culture

Near diploid and normal human mammary epithelial cells (MCF 10A), triple-negative breast cancer cells (MDA-MB-231) and breast cancer cell (MCF-7) were purchased from Procell Life Science & Technology Co., Ltd. (https://www.procell.com.cn/). Normal human colon mucosal epithelial cell (NCM460) and human colon carcinoma cell line (SW620) were purchased from MINGZHOUBIO Co., Ltd. (https://www.mingzhoubio.com/). All cells were cultured with Dulbecco's modified eagle medium (Biological Industries) containing 10% fetal bovine serum (Biological Industries) and incubated at 37˚C in a thermostatic cell incubator containing 5% CO2. Roswell Park Memorial Institute 1640 (Biological Industries) was used to maintain cell growth. All cells retained their original morphology throughout the study period.

Reverse-transcription quantitative PCR (RT-qPCR)

Total RNA from MCF 10A, MDA-MB-231, MCF-7, NCM460 and SW620 was extracted using TRIzol reagent (Mei5 Biotechnology, Co., Ltd., https://mei5bio.com/) according to the manufacturer's instructions and converted into cDNA using M5 Sprint qPCR RT kit with gDNA remover (Mei5 Biotechnology, Co., Ltd.) according to the manufacturer's instructions. The extraction and reverse transcription were performed in an enzyme-free environment. AceQ qPCR SYBR Green Master Mix (Vazyme Biotech Co., Ltd., https://www.vazyme.com/) was used to quantify the relative expression of FADD (Sangon Biotech Co., Ltd.) in mRNA. The primers used were as follows: GAPDH forward, 5'-CAGGAGGCATTGCTGATGAT-3' and reverse, 5'-GAAGGCTGGGGCTCATTT-3'; and FADD forward, 5'-GACCGAGCTCAAGTTCCTATG-3' and reverse, 5'-GAGCATGGAGAAGAGGTCTAG-3'. The thermocycling conditions are provided in Table I.

Table I

Thermocycling conditions of reverse transcription-quantitative PCR.

Table I

Thermocycling conditions of reverse transcription-quantitative PCR.

Stage 1 Pre-denaturationRepeats: 195˚C5 min
Stage 2AmplificationRepeats: 4095˚C10 sec
   60˚C30 sec
Stage 3Melting CurveRepeats: 195˚C15 sec
   60˚C60 sec
   95˚C15 sec
Data acquisition and differential expression of FADD in cancer tissues

Transcriptome data and patient clinical data of 33 human cancers were obtained from TCGA database on the UCSC Xena website (xena.ucsc.edu). All gene names in the expression matrix were transformed from Ensembl ID to the Symbol format. In total, 20 datasets (GSE13057, GSE9750, GSE26566, GSE44076, GSE23400, GSE30784, GSE167093, GSE15641, GSE25097, GSE40791, GSE19188, GSE51024, GSE26712, GSE71729, GSE10927, GSE70770, GSE26253, GSE33630 and GSE63678) containing 2,778 tumor tissues and 1,821 non-tumor tissues were included from the Gene Expression Omnibus (GEO) repository (17-36). The R packages ‘plyr’ (version, 1.8.8; http://cran.ma.ic.ac.uk/web/packages/plyr/plyr.pdf), ‘reshap2’ (version, 1.4.4 http://cran.ma.ic.ac.uk/web/packages/reshape/reshape.pdf) and ‘ggpubr’ (version, 0.6.0; http://cran.ma.ic.ac.uk/web/packages/ggpubr/ggpubr.pdf) were used to create a box plot demonstrating FADD expression differences. Furthermore, the immunohistochemical images of FADD protein in different cancer tissues and normal tissues were obtained from the Human Protein Atlas (HPA; https://www.proteinatlas.org).

FADD alteration and promoter methylation in cancer

FADD alteration data were collected from the cBioPortal website (https://www.cbioportal.org/) for a total of 10,953 patients with cancer, including the corresponding 10,967 samples of mutation and CNA data, for analysis (37). Mutation, structural variant, amplification, deep deletion and multiple alterations of FADD were analyzed in different cancers. The University of Alabama at Birmingham Cancer (UALCAN) data analysis portal (http://ualcan.path.uab.edu) was used to explore differences in promoter methylation levels of FADD between tumor and non-tumor samples in TCGA (38). P<0.05 was considered to indicate a statistically significant difference.

Analysis of survival rate and clinical association of patients with different expressions of FADD

The Kaplan-Meier plotter website (https://kmplot.com) was used to perform overall survival (OS) and relapse-free survival (RFS) prognostic analysis. According to the expression level of FADD, samples were divided into high- and low-expression groups (39). Kaplan Meier analysis was used to compare the differences between OS and RFS between high- and low-expression groups, and values with P<0.05 were considered statistically significant. The Cox proportional hazards model method was used to compare FADD as a continuous variable with survival status and survival time and to calculate the hazard ratio (HR) value and P-value. Values with P<0.05 were considered to indicate a statistically significant difference. A HR value >1 indicated that the expression of FADD was a high-risk factor in the tumor, whereas a value <1 indicated that the expression of FADD was considered. Based on these results, a forest map was created.

Analysis of FADD expression, TME and immune cell infiltration

TME encompasses the internal and external environment in which tumors and tumor cells proliferate, develop and metastasize (40). Changes in TME contribute to the generation of tumor resistance (including immune checkpoint inhibitors resistance) and the metabolic changes in physiological processes (41). The immune infiltration in TME is highly associated with the occurrence and development of tumors and the clinical treatment outcome of patients (42,43). The Spearman correlation test between FADD expression and TME score was performed using the R packages ‘ggplot2’ (version, 3.4.3; https://cran.r-project.org/web/packages/ggplot2/index.html), ‘ggpubr’ and ‘ggExtra’, and the results satisfying the condition (P<0.05, correlation coefficient >0.2) were plotted for visualization. The relative content of immune cells in each sample was determined using the Sangerbox website (http://vip.sangerbox.com/home.html). The relative expression of FADD in the samples and the infiltration of immune cells [B cells, CD4 cells, CD8 cells, neutrophils, macrophages and dendritic cells (DCs)] were analyzed using TIMER2.0 tool (http://timer.cistrome.org/) (44,45).

Correlation of FADD expression with TMB and MSI

Although TMB and MSI (46) are not perfect indicators of cancer immunotherapy response, they are still important biomarkers for predicting the effect of immunotherapy (47,48). The R package ‘fmsb’ (version, 0.7.5; http://cran.ma.ic.ac.uk/web/packages/fmsb/fmsb.pdf) was used to analyze the correlation of the FADD expression with TMB and MSI in all cancer samples (49). P<0.05 was considered to indicate a statistically significant difference. These correlation analysis results were illustrated in a radar map. A correlation coefficient >0 indicated that FADD expression was positively correlated with TMB and MSI, whereas a correlation coefficient <0 indicated that FADD expression was negatively correlated with TMB and MSI.

Gene set enrichment analysis (GSEA)

The GSEA method is useful for the discovery of genes with no significant difference in expression but key biological function (49). Using GSEA website (http://www.gsea-msigdb.org/gsea), data sets were obtained from the Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.kegg.jp/) and Gene Ontology (GO) (http://www.geneology.org) databases. The R packages ‘limma’ (version, 3.56.2; https://bioconductor.org/packages/release/bioc/html/limma.html), ‘org.Hs.eg.db’ (version, 3.17.0; https://bioconductor.org/packages/release/data/annotation/html/org.Hs.eg.db.html), ‘enrichmentplot’ and ‘clusterProfiler’ were used to perform KEGG pathway analysis and GO function annotation analysis on genes differentially expressed between high- and low-expression groups of FADD (49,50). With P<0.05 as the threshold for statistical significance, the top five most significant pathways and biological processes were displayed.

Statistical analysis

FADD expression levels in all cancer tissues and adjacent tissue samples were determined using The R Project for Statistical Computing 4.2.1 (R Foundation and R Core Team, https://www.r-project.org/). The Wilcoxon rank sum test was used to calculate the difference in FADD expression between tumor and non-tumor tissuesand the receiver operating characteristic (ROC) curve was drawn (Sangerbox website, http://vip.sangerbox.com/home.html). A hypothesis test probability (P<0.05) was considered statistically significant. With GAPDH as the internal reference gene, the 2-ΔΔCq method was used to calculate the expression of FADD. Unpaired t-test was used to calculate the significance of the relative expression of FADD between normal breast cells and breast cancer cells. And an unpaired t test with Welch's correction was used to calculate the significance of the relative expression of FADD between colon mucosal epithelial cell and colon carcinoma cell line. Statistical Calculation and Bar Chart Drawing by GraphPad Prism 8.3.0 (Dotmatics).

Results

Expression of FADD in different cancers

The analysis results of the expression of FADD mRNA in tumor and non-tumor tissues collected from the TCGA database revealed that FADD was significantly differentially expressed in 19 cancer types. FADD was relatively highly expressed in bladder urothelial carcinoma (BLCA), BRCA, cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), cholangiocarcinoma (CHOL), colon adenocarcinoma (COAD), ESCA, glioblastoma multiforme (GBM), head and neck squamous cell carcinoma (HNSC), kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), liver hepatocellular carcinoma (LIHC), lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), prostate adenocarcinoma (PRAD), rectum adenocarcinoma (READ), stomach adenocarcinoma STAD, and thyroid carcinoma (THCA) samples, but showed relatively low expression in KICH, and pheochromocytoma and paraganglioma (PCPG) samples (Fig. 1A and B). The results of mRNA expression analysis of FADD in tumor and non-tumor samples collected from the GEO database revealed that FADD was relatively highly expressed in BRCA, CESC, CHOL, COAD, ESCA, KIRC, KIRP, LIHC, LUAD, PAAD and PRAD samples, but showed relatively lower expression in HNSC, mesothelioma (MESO), ovarian serous cystadenocarcinoma (OV), THCA and uterine corpus endometrial carcinoma (UCEC) samples (Fig. 1C and D). RT-qPCR results revealed that FADD mRNA was highly expressed in breast cancer cells and colon cancer cells (Fig. 2A). The results of immunohistochemical analysis of tumor and non-tumor samples in the HPA database showed that FADD protein was relatively highly expressed in breast, colon, liver and gastric cancer tissues (Fig. 2B). Relative expression of FADD in cancer cells is demonstrated in Table II.

Table II

Relative expression of fas-associated death domain in cancer cells.

Table II

Relative expression of fas-associated death domain in cancer cells.

Cell lineMean ± SDP-value
MCF 10A1.1651±0.7826 
MDA-MB-23113.5447±3.93750.0059
MCF-711.4218±4.06220.0127
NCM4601.0059±0.1364 
SW6207.8246±2.44920.0400
FADD alteration in cancer

The analysis results of the types of FADD alteration in 32 cancers in the TCGA database demonstrated that FADD changes were most frequent in patients with ESCA (Fig. 3A). Kaplan-Meier prognosis result of OS in FADD-altered and non-altered patients showed that FADD alteration was significantly associated with shorter OS in cancer patients (Fig. 3B).

FADD promoter methylation level

The analysis of tumor samples and non-tumor samples in the UALCAN database revealed that FADD promoter methylation level was relatively high in CESC (Fig. 4B), ESCA (Fig. 4C), KIRC (Fig. 4D), LUSC (Fig. 4F) and PAAD (Fig. 4G) samples, but lower in BLCA (Fig. 4A), LIHC (Fig. 4E), PRAD (Fig. 4H), sarcoma (SARC; Fig. 4I), testicular germ cell tumors (TGCT; Fig. 4J), THCA (Fig. 4K) and UCEC (Fig. 4L) samples.

Correlation between FADD expression and clinical characteristics of various cancers

To analyze the correlation between the expression of FADD and age, patients were divided into two cohorts: i) Patients aged <65 years and ii) patients aged 65 years or older. The expression level of FADD was relatively higher in elderly cancer patients with ESCA and OV and patients younger than 65 years with skin cutaneous melanoma and TGCT (Fig. 5A). In addition, FADD was highly expressed in female patients with adrenocortical carcinoma and male patients with COAD (Fig. 5B). Notably, the difference in the expression of FADD among different stages of KIRC, LUAD and TGCT patients was statistically significant (Fig. 5C).

Correlation between FADD expression and prognosis of various cancers

The Kaplan-Meier-plot website was used to investigate the relationship between FADD expression and the prognosis of patients with cancer. The results demonstrated that the high expression of FADD was significantly associated with shorter OS in CESC (Fig. 6B), HNSC (Fig. 6C), LIHC (Fig. 6D), LUAD (Fig. 6E), LUSC (Fig. 6F) and PAAD (Fig. 6G), but not in STAD (Fig. 6H), and significantly longer OS in thymoma (THYM) (Fig. 6I) and THCA (Fig. 6J). The low expression of FADD in CESC (Fig. 6L), LUAD (Fig. 6M), PAAD (Fig. 6N) and TGCT (Fig. 6P) was significantly associated with shorter RFS, whereas the high expression of FADD in BRCA (Fig. 6K) and STAD (Fig. 6O) was associated with longer RFS. Cox regression analysis exhibited that FADD was a poor prognostic factor for HNSC, acute myeloid leukemia, brain lower grade glioma (LGG), LIHC, LUAD and PAAD, but a protective factor for MESO and THCA (Fig. 6A).

Effects of FADD on TME and immune cell infiltration

The results of TME analysis showed that the expression of FADD was positively correlated with the ImmuneScore of LGG (Fig. 7A), SARC (Fig. 7B), THCA (Fig. 7D), uterine carcinosarcoma (UCS; Fig. 7E) and uveal melanoma (UVM; Fig. 7F), and negatively correlated with the ImmuneScore of TGCT (Fig. 7C). The expression of FADD was positively correlated with the StromalScore of LGG (Fig. 7G) and negatively correlated with the StromalScore of MESO (Fig. 7H) and THYM (Fig. 7I). Analysis of the data obtained from the TCGA database using the TIMER method revealed that the expression of FADD was positively correlated with B cell infiltration in KIRC, KIPAN, PCPG, PRAD, THYM, THCA, LGG, LIHC, COADREAD, COAD, OV, KICH and ACC, but negatively correlated with B cell infiltration in HNSC, CESC, LUAD and ESCA. Moreover, the expression of FADD was positively correlated with CD4 cell infiltration in KIRC, KIPAN, PCPG, PRAD, LGG, LIHC, COADREAD, COAD, OV, KICH, READ, GBMLGG, GBM and BLCA. The expression of FADD was positively correlated with T cell CD8 infiltration in LIRC, KIPAN, PCPG, PRAD, THYM, THCA, LIHC, COADREAD, COAD, ACC, SKCM-P, GBMLGG, PAAD, BLCA and DLBC, and negatively correlated with T cell CD8 infiltration in HNSC, CESC and GBM. In addition, the expression of FADD was positively correlated with neutrophil and macrophage infiltration in all cancer types except LUSC and DLBC in which the expression of FADD was negatively correlated with macrophage infiltration. Furthermore, the expression of FADD was positively correlated with dendritic cells (DC) infiltration in KIRC, KIPAN, PCPG, PRAD, THYM, THCA, LGG, LIHC, COADREAD, COAD, LUAD, OV, KICH, LUSC, READ, STES, GBMLGG, GBM, KIRP, PAAD, UCEC, BLCA, DLBC and SARC (Fig. 8).

Correlation of FADD expression with MSI and TMB in cancer

FADD was observed to be positively correlated with MSI in LGG, LUAD, PAAD, SARC and UCEC, and negatively correlated with MSI in COAD, PCPG, READ and THYM (Fig. 9A). By contrast, FADD expression was positively correlated with HNSC, KIRC, KIRP, PRAD, SARC, THCA, UCEC and UVM, while it was revealed as negatively correlated with TMB in LUAD and READ (Fig. 9B).

Analysis of different FADD expressions (GSEA)

The expression of FADD was divided into two groups and GSEA analysis was performed. Furthermore, the results demonstrated that FADD was involved in different signaling pathways and biological processes in various cancers. GO enrichment results (Fig. S1) revealed that the main active biological processes associated with the high expression FADD group were the detection of chemical stimulus (in three cancers), epidermal cell differentiation (in three cancers), and epidermis development (in three cancers as well). On the contrary, the main active biological processes associated with the low expression FADD group were the detection of chemical stimulus (in 18 cancers) and the detection of stimulus involved in sensory perception (in 15 cancers). KEGG analysis results (Fig. S2) indicated that the main active signaling pathways in the high expression FADD group were olfactory transduction (in 4 cancers) and systemic lupus atherosclerosis (in 3 cancers). By contrast, the main active signaling pathways associated with the low expression FADD group were olfactory transduction (in 19 cancers) and neuroactive ligand-receptor interaction (in 10 cancers).

Discussion

Analysis of the differential expression of FADD between cancer and normal samples in the TCGA database revealed that FADD was substantially expressed in 18 of the 33 malignancies analyzed and in 11 of the 20 GSE datasets selected. The area under the ROC curve of 6 cancer types (CHOL, GBM, HNSC, LUAD, LUSC and PCPG) from TCGA database had a value of >0.9, and the area under the ROC curve of 4 cancers (ESCA, HNSC, LUAD and STAD) from GEO database had a value of >0.8. RT-qPCR exhibited that FADD mRNA was relatively significantly expressed in breast, colon, liver and stomach cancer cells, which was consistent with immunohistochemical images obtained from the HPA database. These findings showed that FADD may have diagnostic utility as a biomarker for cancer. FADD is a ubiquitous adapter protein that not only conveys apoptotic signals mediated by death receptors but also mediates inflammation and cancer (51-53). Inflammation is a hallmark of a substantial percentage of cancers, which may explain the relatively high expression of FADD in the vast majority of cancers (54,55), including oral squamous cell cancer (56). FADD alteration in the cBioPortal database demonstrated that FADD is more likely to change in more than 30, 20, 10 and 10% of patients with ESCA, HNSC, LUSC and BRCA, respectively, and amplification is the predominant FADD alteration. The human FADD gene is located on chromosome 11q13.3, 11q13-q14 amplification has a relatively high incidence in breast, ovary, head and neck, esophageal, melanoma and bladder tumors, which is consistent with the expression patterns of FADD in cancer (2). This suggests that FADD amplification may cause certain cancers. Methylation of FADD is also linked to cancer. A previous study has identified an association between FADD methylation and oral squamous cell carcinoma (57). The UALCAN database analysis revealed that abnormal FADD promoter methylation was associated with 12 tumor samples, indicating that both FADD mutation and promoter methylation are associated with malignancy. Compared with normal tissue, FADD promoter methylation levels were significantly reduced in primary tumors of BLCA, LIHC, PRAD and THCA, and differential analysis revealed that FADD mRNA was significantly highly expressed in these cancer tissues. The high expression of FADD mRNA in BLCA, LIHC, PRAD and THCA may be related to the decrease of the promoter methylation level. FADD mRNA was also highly expressed in CESC, KIRC and LUSC, but FADD promoter methylation levels were significantly lower in primary tumor tissues than in corresponding normal tissues in these cancers. This may be due to the low expression or no expression of FADD mRNA in the corresponding normal tissue. Even if the methylation level of the promoter is increased, the expression of FADD in primary tissue remains significantly higher than that in normal tissue. This suggests that FADD is reliable as a biomarker for the diagnosis of these cancers.

High expression of FADD was significantly associated with shorter OS in six types of cancer patients and RFS in four types of cancer patients, as exhibited by Kaplan-Meier analysis. FADD expression was a risk factor for numerous cancers (6 types) and a protective factor for fewer cancers (2 types), according to Cox regression analysis. For instance, Kaplan-Meier prognostic analysis and Cox regression analysis of patients with HNSC, LIHC and LUAD revealed that FADD expression was a risk factor for these malignancies. A recent study has demonstrated the predictive utility of FADD gene can in the prognosis of lung adenocarcinoma in women (58). Because FADD amplification occurs in high frequency in HNSC, numerous studies have investigated its potential as a biomarker of HNSC (59,60). Additionally, the immunohistochemical results of FADD overexpression were substantially linked with poor OS in patients with HNSC, according to a previous meta-analysis (61). As one of the apoptosis-related factors, FADD is associated with the occurrence of LIHC, but further research is needed to confirm its prognostic value for patients with LIHC (62-64). Different prognostic analysis approaches have demonstrated that FADD predicts poor OS in LIHC patients, indicating that FADD may be employed as both a diagnostic and prognostic biomarker for patients with LIHC. Analysis of TME and immune cell infiltration revealed that FADD expression influences tumor immunity in a number of malignancies, particularly various tumors where neutrophil and DC infiltration are strongly positively associated. The neutrophil is a crucial cell that regulates inflammation and immune response, whereas DC is an antigen-presenting cell with a significant effect on tumor immunity (65-68). This suggests that FADD may influence tumor immunity by boosting neutrophil and DC infiltration into tumors. FADD expression was substantially related to MSI in 9 malignancies and TMB in 10 tumors, suggesting its potential as an immunotherapy marker. Bowman et al (69) revealed that the phosphorylation of FADD promoted the proliferation of lung cancer cells, suggesting that FADD indeed plays a role in tumorigenesis and development, and it is necessary to conduct in-depth research on it in the future.

Because of inconsistencies between the GEO and TCGA databases, expression data for 33 tumors was not gathered to verify the differential expression of FADD. The present study is limited to the expression and clinical relevance of FADD in different cancers, and lacks clarification of the specific role of FADD in tumorigenesis and progression, which is necessary to explore FADD as a therapeutic target. Although FADD expression was identified as a potential diagnostic and prognostic biomarker for specific cancers, its clinical application and applicability in clinical practice need to be rigorously evaluated and verified by large-scale clinical trials.

The present study carefully evaluated the expression of FADD in various malignancies and its effect on the prognoses of patients with cancer. Analysis of various databases revealed that FADD was highly expressed in BRCA, CESC, CHOL, COAD, ESCA, KIRC, KIRP, LIHC, LUAD and PRAD. Moreover, the high expression of FADD was confirmed in BRCA, COAD, LIHC and STAD using RT-qPCR, supporting the potential utility of FADD as a prognostic biomarker for patients with LIHC. In conclusion, FADD is highly expressed in numerous malignancies and can be utilized as a diagnostic biomarker for BRCA, COAD, LIHC, and STAD. FADD expression is a predictive risk factor for HNSC, LIHC, and LUAD patients and has potential value as a prognostic marker for these tumors.

Supplementary Material

GO enrichment analysis of differential genes in high and low expression groups of FADD. GO, Gene Ontology; FADD, fas-associated death domain; GOBP, GO biological process; DLBC, lymphoid neoplasm diffuse large B-cell lymphoma; ESCA, esophageal carcinoma; GBM, glioblastoma multiforme; HNSC, head and neck squamous cell carcinoma; KICH, kidney chromophobe; KIRC, kidney renal clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LAML, acute myeloid leukemia; LGG, brain lower grade glioma; LIHC, liver hepatocellular carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; MESO, mesothelioma; OV, ovarian serous cystadenocarcinoma; PAAD, pancreatic adenocarcinoma; PCPG, pheochromocytoma and paraganglioma; PRAD, prostate adenocarcinoma; READ, rectum adenocarcinoma; SARC, sarcoma; SKCM, skin cutaneous melanoma; STAD, stomach adenocarcinoma; TGCT, testicular germ cell tumors; THCA, thyroid carcinoma; THYM, thymoma; UCEC, uterine corpus endometrial carcinoma; UCS, uterine carcinosarcoma; UVM, uveal melanoma; AAC, adenoid cystic carcinoma; BLCA, bladder urothelial carcinoma; BRCA, breast invasive carcinoma; CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma; CHOL, cholangiocarcinoma; COAD, colon adenocarcinoma.
KEGG enrichment analysis of differential genes in high and low expression groups of FADD. KEGG, Kyoto Encyclopedia of Genes and Genomes; FADD, fas-associated death domain; PRAD, prostate adenocarcinoma; PCPG, pheochromocytoma and paraganglioma; PAAD, pancreatic adenocarcinoma; OV, ovarian serous cystadenocarcinoma; LUSC, lung squamous cell carcinoma; LUAD, lung adenocarcinoma; LIHC, liver hepatocellular carcinoma; LGG, brain lower grade glioma; READ, rectum adenocarcinoma; SARC, sarcoma; SKCM, skin cutaneous melanoma; STAD, stomach adenocarcinoma; TGCT, testicular germ cell tumors; THCA, thyroid carcinoma; THYM, thymoma; UCEC, uterine corpus endometrial carcinoma; BRCA, breast invasive carcinoma; CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma; CHOL, cholangiocarcinoma; COAD, colon adenocarcinoma; DLBC, lymphoid neoplasm diffuse large B-cell lymphoma; ESCA, esophageal carcinoma; GBM, glioblastoma multiforme; HNSC, head and neck squamous cell carcinoma; KICH, kidney chromophobe; KIRC, kidney renal clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LAML, acute myeloid leukemia.

Acknowledgements

The authors wish to thank the Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province and the DaVinci Surgery System Database (www.davincisurgery.com) for their help and support in the methodology.

Funding

Funding: The present study was supported by the 2021 Central to guide local scientific and Technological Development (grant no. ZYYDDFFZZJ-1), the Natural Science Foundation of Gansu Province, China (grant no. 18JR3RA052), the Lanzhou Talent Innovation and Entrepreneurship Project Task Contract (grant no. 2016-RC-56), the Gansu Da Vinci robot high-end diagnosis and treatment team construction project, the Gansu Provincial Youth Science and Technology Fund Program (grant no. 20JR10RA415) and the National Key Research and Development Program (grant no. 2018YFC1311500).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in TCGA (https://portal.gdc.cancer.gov/), UCSC Xena website (xena.ucsc.edu/), Kaplan Meier plotter portal (https://kmplot.com/), GEO database (https://www.ncbi.nlm.nih.gov/), The Molecular Signatures Database (https://www.gsea-msigdb.org/gsea/msigdb), The Human Protein Atlas (https://www.proteinatlas.org/), cBioPortal database (http://www.cbioportal.org/), UALCAN portal (ualcan.path.uab.edu/) and sangerbox website (http://vip.sangerbox.com/home.html).

Authors' contributions

XJ and CW conceived the study. ZX and QZ comprehensively collected relevant data. XJ and CW contributed in data analysis and in drafting the manuscript. XJ and CW confirm the authenticity of all the raw data. CW revised the manuscript and HC reviewed the manuscript. All authors read and approved the final version of the manuscript.

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 

Mouasni S and Tourneur L: FADD at the crossroads between cancer and inflammation. Trends Immunol. 39:1036–1053. 2018.PubMed/NCBI View Article : Google Scholar

2 

Wilkerson PM and Reis-Filho JS: The 11q13-q14 amplicon: Clinicopathological correlations and potential drivers. Genes Chromosomes Cancer. 52:333–355. 2013.PubMed/NCBI View Article : Google Scholar

3 

Saggioro FP, Neder L, Stávale JN, Paixão-Becker AN, Malheiros SM, Soares FA, Pittella JE, Matias CC, Colli BO, Carlotti CG Jr and Franco M: Fas, FasL, and cleaved caspases 8 and 3 in glioblastomas: A tissue microarray-based study. Pathol Res Pract. 210:267–273. 2014.PubMed/NCBI View Article : Google Scholar

4 

Marín-Rubio JL, Vela-Martín L, Fernández-Piqueras J and Villa-Morales M: FADD in cancer: Mechanisms of altered expression and function, and clinical implications. Cancers (Basel). 11(1462)2019.PubMed/NCBI View Article : Google Scholar

5 

Tourneur L and Chiocchia G: FADD: A regulator of life and death. Trends Immunol. 31:260–269. 2010.PubMed/NCBI View Article : Google Scholar

6 

Zhuang H, Gan Z, Jiang W, Zhang X and Hua ZC: Functional specific roles of FADD: Comparative proteomic analyses from knockout cell lines. Mol Biosyst. 9:2063–2078. 2013.PubMed/NCBI View Article : Google Scholar

7 

Screaton RA, Kiessling S, Sansom OJ, Millar CB, Maddison K, Bird A, Clarke AR and Frisch SM: Fas-associated death domain protein interacts with methyl-CpG binding domain protein 4: A potential link between genome surveillance and apoptosis. Proc Natl Acad Sci USA. 100:5211–5216. 2003.PubMed/NCBI View Article : Google Scholar

8 

Gómez-Angelats M and Cidlowski JA: Molecular evidence for the nuclear localization of FADD. Cell Death Differ. 10:791–797. 2003.PubMed/NCBI View Article : Google Scholar

9 

Zhang Y and Zhang Z: The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 17:807–821. 2020.PubMed/NCBI View Article : Google Scholar

10 

Jiang Y, Chen M, Nie H and Yuan Y: PD-1 and PD-L1 in cancer immunotherapy: Clinical implications and future considerations. Hum Vaccin Immunother. 15:1111–1122. 2019.PubMed/NCBI View Article : Google Scholar

11 

Gajewski TF, Schreiber H and Fu YX: Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 14:1014–1022. 2013.PubMed/NCBI View Article : Google Scholar

12 

Balkwill FR, Capasso M and Hagemann T: The tumor microenvironment at a glance. J Cell Sci. 125:5591–5596. 2012.PubMed/NCBI View Article : Google Scholar

13 

Mellman I, Coukos G and Dranoff G: Cancer immunotherapy comes of age. Nature. 480:480–489. 2011.PubMed/NCBI View Article : Google Scholar

14 

Esfahani K, Roudaia L, Buhlaiga N, Del Rincon SV, Papneja N and Miller WH*Jr: A review of cancer immunotherapy: From the past, to the present, to the future. Curr Oncol. 27 (Suppl 2):S87–S97. 2020.PubMed/NCBI View Article : Google Scholar

15 

Ranjan K, Waghela BN, Vaidya FU and Pathak C: Cell-penetrable peptide-conjugated FADD induces apoptosis and regulates inflammatory signaling in cancer cells. Int J Mol Sci. 21(6890)2020.PubMed/NCBI View Article : Google Scholar

16 

Xiang R, Liu Y, Zhu L, Dong W and Qi Y: Adaptor FADD is recruited by RTN3/HAP in ER-bound signaling complexes. Apoptosis. 11:1923–1932. 2006.PubMed/NCBI View Article : Google Scholar

17 

Kim WJ, Kim EJ, Kim SK, Kim YJ, Ha YS, Jeong P, Kim MJ, Yun SJ, Lee KM, Moon SK, et al: Predictive value of progression-related gene classifier in primary non-muscle invasive bladder cancer. Mol Cancer. 9(3)2010.PubMed/NCBI View Article : Google Scholar

18 

Wang J, Zhang X, Beck AH, Collins LC, Collins LC, Chen WY, Tamimi RM, Hazra A, Brown M, Rosner B and Hankinson SE: Alcohol consumption and risk of breast cancer by tumor receptor expression. Horm Cancer. 6:237–246. 2015.PubMed/NCBI View Article : Google Scholar

19 

Scotto L, Narayan G, Nandula SV, Arias-Pulido H, Subramaniyam S, Schneider A, Kaufmann AM, Wright JD, Pothuri B, Mansukhani M and Murty VV: Identification of copy number gain and overexpressed genes on chromosome arm 20q by an integrative genomic approach in cervical cancer: Potential role in progression. Genes Chromosomes Cancer. 47:755–765. 2008.PubMed/NCBI View Article : Google Scholar

20 

Andersen JB, Spee B, Blechacz BR, Avital I, Komuta M, Barbour A, Conner EA, Gillen MC, Roskams T, Roberts LR, et al: Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology. 142:1021–1031.e15. 2012.PubMed/NCBI View Article : Google Scholar

21 

Solé X, Crous-Bou M, Cordero D, Olivares D, Guinó E, Sanz-Pamplona R, Rodriguez-Moranta F, Sanjuan X, de Oca J, Salazar R and Moreno V: Discovery and validation of new potential biomarkers for early detection of colon cancer. PLoS One. 9(e106748)2014.PubMed/NCBI View Article : Google Scholar

22 

Su H, Hu N, Yang HH, Wang C, Takikita M, Wang QH, Giffen C, Clifford R, Hewitt SM, Shou JZ, et al: Global gene expression profiling and validation in esophageal squamous cell carcinoma and its association with clinical phenotypes. Clin Cancer Res. 17:2955–2966. 2011.PubMed/NCBI View Article : Google Scholar

23 

Chen C, Méndez E, Houck J, Fan W, Lohavanichbutr P, Doody D, Yueh B, Futran ND, Upton M, Farwell DG, et al: Gene expression profiling identifies genes predictive of oral squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 17:2152–2162. 2008.PubMed/NCBI View Article : Google Scholar

24 

Laskar RS, Li P, Ecsedi S, Abedi-Ardekani B, Durand G, Robinot N, Hubert JN, Janout V, Zaridze D, Mukeria A, et al: Sexual dimorphism in cancer: Insights from transcriptional signatures in kidney tissue and renal cell carcinoma. Hum Mol Genet. 30:343–355. 2021.PubMed/NCBI View Article : Google Scholar

25 

Jones J, Otu H, Spentzos D, Kolia S, Inan M, Beecken WD, Fellbaum C, Gu X, Joseph M, Pantuck AJ, et al: Gene signatures of progression and metastasis in renal cell cancer. Clin Cancer Res. 11:5730–5739. 2005.PubMed/NCBI View Article : Google Scholar

26 

Ivanovska I, Zhang C, Liu AM, Wong KF, Lee NP, Lewis P, Philippar U, Bansal D, Buser C, Scott M, et al: Gene signatures derived from a c-MET-driven liver cancer mouse model predict survival of patients with hepatocellular carcinoma. PLoS One. 6(e24582)2011.PubMed/NCBI View Article : Google Scholar

27 

Zhang Y, Foreman O, Wigle DA, Kosari F, Vasmatzis G, Salisbury JL, van Deursen J and Galardy PJ: USP44 regulates centrosome positioning to prevent aneuploidy and suppress tumorigenesis. J Clin Invest. 122:4362–4374. 2012.PubMed/NCBI View Article : Google Scholar

28 

Hou J, Aerts J, den Hamer B, van Ijcken W, den Bakker M, Riegman P, van der Leest C, van der Spek P, Foekens JA, Hoogsteden HC, et al: Gene expression-based classification of non-small cell lung carcinomas and survival prediction. PLoS One. 5(e10312)2010.PubMed/NCBI View Article : Google Scholar

29 

Suraokar MB, Nunez MI, Diao L, Chow CW, Kim D, Behrens C, Lin H, Lee S, Raso G, Moran C, et al: Expression profiling stratifies mesothelioma tumors and signifies deregulation of spindle checkpoint pathway and microtubule network with therapeutic implications. Ann Oncol. 25:1184–1192. 2014.PubMed/NCBI View Article : Google Scholar

30 

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.PubMed/NCBI View Article : Google Scholar

31 

Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, Rashid NU, Williams LA, Eaton SC, Chung AH, et al: Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 47:1168–1178. 2015.PubMed/NCBI View Article : Google Scholar

32 

Giordano TJ, Kuick R, Else T, Gauger PG, Vinco M, Bauersfeld J, Sanders D, Thomas DG, Doherty G and Hammer G: Molecular classification and prognostication of adrenocortical tumors by transcriptome profiling. Clin Cancer Res. 15:668–676. 2009.PubMed/NCBI View Article : Google Scholar

33 

Whitington T, Gao P, Song W, Ross-Adams H, Lamb AD, Yang Y, Svezia I, Klevebring D, Mills IG, Karlsson R, et al: Gene regulatory mechanisms underpinning prostate cancer susceptibility. Nat Genet. 48:387–397. 2016.PubMed/NCBI View Article : Google Scholar

34 

Oh SC, Sohn BH, Cheong JH, Kim SB, Lee JE, Park KC, Lee SH, Park JL, Park YY, Lee HS, et al: Clinical and genomic landscape of gastric cancer with a mesenchymal phenotype. Nat Commun. 9(1777)2018.PubMed/NCBI View Article : Google Scholar

35 

Dom G, Tarabichi M, Unger K, Thomas G, Oczko-Wojciechowska M, Bogdanova T, Jarzab B, Dumont JE, Detours V and Maenhaut C: A gene expression signature distinguishes normal tissues of sporadic and radiation-induced papillary thyroid carcinomas. Br J Cancer. 107:994–1000. 2012.PubMed/NCBI View Article : Google Scholar

36 

Pappa KI, Polyzos A, Jacob-Hirsch J, Amariglio N, Vlachos GD, Loutradis D and Anagnou NP: Profiling of discrete gynecological cancers reveals novel transcriptional modules and common features shared by other cancer types and embryonic stem cells. PLoS One. 10(e0142229)2015.PubMed/NCBI View Article : Google Scholar

37 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012.PubMed/NCBI View Article : Google Scholar

38 

Zhang Y, Chen F, Chandrashekar DS, Varambally S and Creighton CJ: Proteogenomic characterization of 2002 human cancers reveals pan-cancer molecular subtypes and associated pathways. Nat Commun. 13(2669)2022.PubMed/NCBI View Article : Google Scholar

39 

Lánczky A and Győrffy B: Web-based survival analysis tool tailored for medical research (KMplot): Development and implementation. J Med Internet Res. 23(e27633)2021.PubMed/NCBI View Article : Google Scholar

40 

Whiteside TJ: The tumor microenvironment and its role in promoting tumor growth. Oncogene. 27:5904–5912. 2008.PubMed/NCBI View Article : Google Scholar

41 

Arneth B: Tumor microenvironment. Medicina (Kaunas). 56(15)2019.PubMed/NCBI View Article : Google Scholar

42 

Fridman WH, Galon J, Dieu-Nosjean MC, Cremer I, Fisson S, Damotte D, Pagès F, Tartour E and Sautès-Fridman C: Immune infiltration in human cancer: Prognostic significance and disease control. Curr Top Microbiol Immunol. 344:1–24. 2011.PubMed/NCBI View Article : Google Scholar

43 

Pagès F, Galon J, Dieu-Nosjean MC, Tartour E, Sautès-Fridman C and Fridman WH: Immune infiltration in human tumors: A prognostic factor that should not be ignored. Oncogene. 29:1093–1102. 2010.PubMed/NCBI View Article : Google Scholar

44 

Shen W, Song Z, Zhong X, Huang M, Shen D, Gao P, Qian X, Wang M, He X, Wang T, et al: Sangerbox: A comprehensive, interaction-friendly clinical bioinformatics analysis platform. iMeta. 1(e36)2022.

45 

Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, Li B and Liu XS: TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 48 (W1):W509–W514. 2020.PubMed/NCBI View Article : Google Scholar

46 

Niu B, Ye K, Zhang Q, Lu C, Xie M, McLellan MD, Wendl MC and Ding L: MSIsensor: Microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics. 30:1015–1016. 2014.PubMed/NCBI View Article : Google Scholar

47 

Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, Stephens PJ, Daniels GA and Kurzrock R: Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 16:2598–2608. 2017.PubMed/NCBI View Article : Google Scholar

48 

Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, Schrock A, Campbell B, Shlien A, Chmielecki J, et al: Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 9(34)2017.PubMed/NCBI View Article : Google Scholar

49 

Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 34:267–273. 2003.PubMed/NCBI View Article : Google Scholar

50 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000.PubMed/NCBI View Article : Google Scholar

51 

Harris MA, Clark J, Ireland A, Lomax J, Ashburner M, Foulger R, Eilbeck K, Lewis S, Marshall B, Mungall C, et al: The gene ontology (GO) database and informatics resource. Nucleic Acids Res. 32 (Database Issue):D258–D261. 2004.PubMed/NCBI View Article : Google Scholar

52 

Sharma VK, Singh TG, Singh S, Garg N and Dhiman S: Apoptotic pathways and Alzheimer's disease: Probing therapeutic potential. Neurochem Res. 46:3103–3122. 2021.PubMed/NCBI View Article : Google Scholar

53 

Zhou W, Lai Y, Zhu J, Xu X, Yu W, Du Z, Wu L, Zhang X and Hua Z: The classical apoptotic adaptor FADD regulates glycolytic capacity in acute lymphoblastic leukemia. Int J Biol Sci. 18:3137–3155. 2022.PubMed/NCBI View Article : Google Scholar

54 

Lee EW, Seo J, Jeong M, Lee S and Song J: The roles of FADD in extrinsic apoptosis and necroptosis. BMB Rep. 45:496–508. 2012.PubMed/NCBI View Article : Google Scholar

55 

Singh N, Baby D, Rajguru JP, Patil PB, Thakkannavar SS and Pujari VB: Inflammation and cancer. Ann Afr Med. 18:121–126. 2019.PubMed/NCBI View Article : Google Scholar

56 

Murata M: Inflammation and cancer. Environ Health Prev Med. 23(50)2018.PubMed/NCBI View Article : Google Scholar

57 

Prapinjumrune C, Morita K, Kuribayashi Y, Hanabata Y, Shi Q, Nakajima Y, Inazawa J and Omura K: DNA amplification and expression of FADD in oral squamous cell carcinoma. J Oral Pathol Med. 39:525–532. 2010.PubMed/NCBI View Article : Google Scholar

58 

Saberi E, Kordi-Tamandani DM, Jamali S and Rigi-Ladiz MA: Analysis of methylation and mRNA expression status of FADD and FAS genes in patients with oral squamous cell carcinoma. Med Oral Patol Oral Cir Bucal. 19:e562–e568. 2014.PubMed/NCBI View Article : Google Scholar

59 

Liu Z, Zhang K, Zhao Z, Qin Z and Tang H: Prognosis-related autophagy genes in female lung adenocarcinoma. Medicine (Baltimore). 101(e28500)2022.PubMed/NCBI View Article : Google Scholar

60 

Jiang Y, Li Y, Ge H, Wu Y, Zhang Y, Guo S, Zhang P, Cheng J and Wang Y: Identification of an autophagy-related prognostic signature in head and neck squamous cell carcinoma. J Oral Pathol Med. 50:1040–1049. 2021.PubMed/NCBI View Article : Google Scholar

61 

Fan S, Müller S, Chen ZG, Pan L, Tighiouart M, Shin DM, Khuri FR and Sun SY: Prognostic impact of Fas-associated death domain, a key component in death receptor signaling, is dependent on the presence of lymph node metastasis in head and neck squamous cell carcinoma. Cancer Biol Ther. 14:365–369. 2013.PubMed/NCBI View Article : Google Scholar

62 

González-Moles MÁ, Ayén Á, González-Ruiz I, de Porras-Carrique T, González-Ruiz L, Ruiz-Ávila I and Ramos-García P: Prognostic and clinicopathological significance of FADD upregulation in head and neck squamous cell carcinoma: A systematic review and meta-analysis. Cancers (Basel). 12(2393)2020.PubMed/NCBI View Article : Google Scholar

63 

Verboom L, Martens A, Priem D, Hoste E, Sze M, Vikkula H, Van Hove L, Voet S, Roels J, Maelfait J, et al: OTULIN prevents liver inflammation and hepatocellular carcinoma by inhibiting FADD- and RIPK1 kinase-mediated hepatocyte apoptosis. Cell Rep. 30:2237–2247.e6. 2020.PubMed/NCBI View Article : Google Scholar

64 

Harari-Steinfeld R, Gefen M, Simerzin A, Zorde-Khvalevsky E, Rivkin M, Ella E, Friehmann T, Gerlic M, Zucman-Rossi J, Caruso S, et al: The lncRNA H19-derived MicroRNA-675 promotes liver necroptosis by targeting FADD. Cancers (Basel). 13(411)2021.PubMed/NCBI View Article : Google Scholar

65 

Liu W, Jing ZT, Xue CR, Wu SX, Chen WN, Lin XJ and Lin X: PI3K/AKT inhibitors aggravate death receptor-mediated hepatocyte apoptosis and liver injury. Toxicol Appl Pharmacol. 381(114729)2019.PubMed/NCBI View Article : Google Scholar

66 

Euler M and Hoffmann MH: The double-edged role of neutrophil extracellular traps in inflammation. Biochem Soc Trans. 47:1921–1930. 2019.PubMed/NCBI View Article : Google Scholar

67 

Liew PX and Kubes P: The neutrophil's role during health and disease. Physiol Rev. 99:1223–1248. 2019.PubMed/NCBI View Article : Google Scholar

68 

Murphy TL and Murphy KM: Dendritic cells in cancer immunology. Cell Mol Immunol. 19:3–13. 2022.PubMed/NCBI View Article : Google Scholar

69 

Bowman BM, Sebolt KA, Hoff BA, Boes JL, Daniels DL, Heist KA, Galbán CJ, Patel RM, Zhang J, Beer DG, et al: Phosphorylation of FADD by the kinase CK1α promotes KRASG12D-induced lung cancer. Sci Signal. 8(ra9)2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2023
Volume 19 Issue 5

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang C, Jiang X, Zhao Q, Xie Z and Cai H: The diagnostic or prognostic values of FADD in cancers based on pan‑cancer analysis. Biomed Rep 19: 77, 2023
APA
Wang, C., Jiang, X., Zhao, Q., Xie, Z., & Cai, H. (2023). The diagnostic or prognostic values of FADD in cancers based on pan‑cancer analysis. Biomedical Reports, 19, 77. https://doi.org/10.3892/br.2023.1659
MLA
Wang, C., Jiang, X., Zhao, Q., Xie, Z., Cai, H."The diagnostic or prognostic values of FADD in cancers based on pan‑cancer analysis". Biomedical Reports 19.5 (2023): 77.
Chicago
Wang, C., Jiang, X., Zhao, Q., Xie, Z., Cai, H."The diagnostic or prognostic values of FADD in cancers based on pan‑cancer analysis". Biomedical Reports 19, no. 5 (2023): 77. https://doi.org/10.3892/br.2023.1659