Next Article in Journal
Multi-Stress Accelerated Degradation Testing Reliability Assessment of LED Lamp Beads Considering Generalized Coupling
Previous Article in Journal
DCW-YOLO: An Improved Method for Surface Damage Detection of Wind Turbine Blades
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Signaling Pathways in Clear Cell Renal Cell Carcinoma and Candidate Drugs Unveiled through Transcriptomic Network Analysis of Hub Genes

1
School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila City 1002, Philippines
2
School of Graduate Studies, Mapúa University, Manila City 1002, Philippines
3
Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan
4
Department of Biology, School of Health Sciences, Mapúa University, Makati City 1002, Philippines
*
Author to whom correspondence should be addressed.
Appl. Sci. 2024, 14(19), 8768; https://doi.org/10.3390/app14198768 (registering DOI)
Submission received: 13 August 2024 / Revised: 12 September 2024 / Accepted: 23 September 2024 / Published: 28 September 2024
(This article belongs to the Section Biomedical Engineering)

Abstract

:
Clear cell renal cell carcinoma (ccRCC) is a type of kidney cancer. It advances quickly and often metastasizes, making the prognosis for patients challenging. This study used weighted gene co-expression network analysis (WGCNA) to study gene expression data of different stages of ccRCC obtained in the GEO database. The analysis identified three significant highly preserved gene modules across the datasets: GSE53757, GSE22541, GSE66272, and GSE73731. Functional annotation and pathway enrichment analysis using DAVID revealed inflammatory pathways (e.g., NF-kB, Hippo, and HIF-1 pathways) that may drive ccRCC development and progression. The study also introduced the involvement of viral infections associated with the disease in the metabolic reprogramming of ccRCC. A drug repurposing analysis was also conducted to identify potential drug candidates for ccRCC using the upregulated and downregulated hub genes. The top candidates are ziprasidone (dopamine and serotonin receptor antagonist) and fentiazac (cyclooxygenase inhibitor). Other drug candidates were also obtained, such as phosphodiesterase/DNA methyltransferase/ATM kinase inhibitors, acetylcholine antagonists, and NAD precursors. Overall, the study’s findings suggest that identifying several genes and signaling pathways related to ccRCC may uncover new targets, biomarkers, and even drugs that can be repurposed, which can help develop new and effective treatments for the disease.

1. Introduction

Renal cell carcinoma (RCC) accounts for 3% of all cancers worldwide. The clear cell type is the most common RCC. It is responsible for 80% of all RCCs [1,2]. A clear cell appearance of the tumor cells, caused by the accumulation of lipids and glycogen in the cytoplasm, is the characteristic feature of ccRCC [3]. RCC is categorized according to the TNM classification system. The cancer stages are categorized in terms of tumor size, degree of lymph nodes, and metastasis [4]. In the cases of RCC, both Stage 1 (≤7 cm) and 2 tumors (≥7 cm) are still confined in the kidney. On the other hand, Stage 3 has already spread to the nearby parts of the kidney or lymph nodes, and Stage 4 indicates metastasis to distant sites [5,6,7,8,9]. In terms of survival rates of patients, stage 1 has 90.4%, stage 2 has 83.4%, stage 3 has 66.0%, and the rate is 9.1% for stage 4 [6,7]. The development of ccRCC mutations or loss of the VHL gene disrupts the oxygen regulation response, which accumulates hypoxia-inducible factors such as HIF-1α and HIF-2α. This, in turn, activates certain genes like vascular endothelial growth factor (VEGF), which contribute to the physiology of the tumor and promote cancer [10,11,12,13]. Inflammation may also worsen the ccRCC [14,15,16,17] since significant leukocyte infiltrates, including CD8+ and CD4+ T cells, are linked with higher tumor grade and poor prognosis in ccRCC [14,18].
The early stages of ccRCC are usually managed with surgery or conventional treatments. Still, as the disease progresses to a later stage, the treatment becomes more complex and requires multiple therapies with a combination of tyrosine kinase/immune checkpoint/mTOR inhibitors [19,20]. Along with that, drug resistance and severe side effects are also considered problems in treating ccRCC [3,20,21,22]. Currently, researchers are exploring how combining traditional treatments with new approaches might address these problems. However, it has been found that simply adding existing therapies together only resolves some of the issues [20]. That is why new treatments are needed. Studies have shown also that treatment responses can vary due to many factors (e.g., genetic differences, inflammation). This emphasizes the need for a deeper understanding of ccRCC [3].
To address these challenges, bioinformatics techniques such as weighted gene co-expression network analysis (WGCNA) can be used. WGCNA analyzes the relationship of genes in line with their expression patterns across different diseases or stages/grades of a particular disease. It helps identify important gene groups that can be analyzed to determine key genes that are significant or related to the disease’s development, progression, and even pathways associated with the disease [23]. By identifying hub/key genes, researchers can also use this to identify existing drugs that can be repurposed based on their gene signatures and patterns. These genes may also uncover significant pathways that can reveal therapeutic opportunities. Recent studies employing WGCNA have explored diseases [24,25] including cancer. This suggests that bioinformatics analysis may lead to discovering new biomarkers and important pathways involved in the disease [24,25,26].
This study used four gene expression datasets (GSE53757, GSE22541, GSE66272, and GSE73731) to analyze different stages of ccRCC. Bioinformatics tools such as WGCNA, DAVID, STRING, and Cytoscape were used to identify gene modules. These gene modules are used to determine functional annotations, pathways, and key genes associated with ccRCC. The identified hub genes were also used to facilitate the repurposing of existing drugs through drug repurposing analysis.

2. Materials and Methods

2.1. Data Retrieval and Preliminary Processing

Gene expression data from DNA microarray analyses of tumor samples from patients with ccRCC stages 1 to 4 were obtained from the Gene Expression Omnibus (GEO) of the National Center for Biotechnology Information (NCBI) [https://www.ncbi.nlm.nih.gov/geo/, accessed on 12 June 2024]. Only datasets pre-processed using the GPL570-HG-U133 Plus 2 Affymetrix Human Genome U133 Plus 2.0 Array were selected and used to ensure consistency and avoid/minimize variations from probe design or normalization differences [24]. The study analyzed 113 samples: 24 from stage 1 (S1), 21 from stage 2 (S2), 27 from stage 3 (S3), and 44 from stage 4 (S4). Detailed information regarding the samples is provided in Table 1.
The datasets were processed in R (v.4.4.0) using the affy package from Bioconductor v3.18 (www.bioconductor.org, accessed on 27 June 2024). This package performed background correction, quantile normalization, and log-2 transformation of the data. The outliers were identified through boxplots and dendrograms. The control probes were removed to reduce variation. Gene expression data were also filtered for genes above the 20th percentile. Probes in all datasets were retained, and samples lacking values after log-2 transformation were removed using the goodSamplesGenesMs function of the WGCNA R package. The probe IDs were converted to gene symbols with the AnnotationDbi function and the hgu133plus2.db [24].

2.2. Weighted Gene Co-Expression Network Analysis (WGCNA)

2.2.1. Determining the Optimal Soft-Thresholding Power for Scale-Free Network

Scale-free topology fit against the power index (1 to 20) was calculated using the pickSoftThreshold function in the WGCNA package in R. The soft-threshold power (β) was selected as the value that met the scale-free topology criterion. This analyzes gene degree distributions using power-law distributions. This assessment also includes checking the network connectivity with linear patterns on a log–log scale. Additionally, a plot of scale-free topology fit against β was used to identify the power at which the fit stabilized or was sufficiently high. To vary the chosen β, an approximate straight-line relationship was generated and examined using soft-connectivity (k) values [27].

2.2.2. Construction of Networks and Identification of Modules

The adjacency matrices for network construction were created using Pearson’s correlation with the “signed” network type. Topological overlap measure (TOM) dissimilarities were computed by raising the adjacency matrices to β. This emphasizes strong correlations and downweighs weak ones. Gene clustering was performed hierarchically using the flashClust function. Dendrograms were generated using the hclust function and the “average” method. Gene modules were identified using cuttreeHybrid using deep split parameters from 0 to 3 [23]. Stable modules across different parameters suggest good clustering, while varying results indicate a need for further validation.

2.3. Module Preservation Analysis

Module preservation analysis measures how well gene group/module connectivity patterns are maintained across different networks and conditions. It determines whether these gene groups remain biologically significant across several diseases or conditions. Module preservation analysis was carried out using the modulePreservation function of the WGCNA package on ccRCC S1 to S4 datasets. The parameters used are “signed” network type, 1000 permutations, and a minimum module size of 100. Eigengene-based connectivity (kME) was computed with the moduleEigengenes function to identify module genes based on their correlation with module eigengenes [23].

2.4. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathways Analysis

Genes obtained from each preserved module were subjected to functional and pathway enrichment analysis using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) (https://david.ncifcrf.gov/home.jsp, accessed on 28 June 2024). Gene Ontology (GO) analysis was carried out to identify biological processes (BP), molecular functions (MF), and cellular components associated with ccRCC. The KEGG database was also used to determine important pathways. Results with a p-value of less than 0.05 were considered statically significant and were considered for further analysis.

2.5. Protein–Protein Interaction (PPI) Network Analysis

The networks of each module were constructed using a high confidence level score of 0.7 in STRING v 12.0 (https://string-db.org/, accessed on 6 September 2024). Following that, their screened PPI networks were imported into Cytoscape v3.10.1 for further analysis. Topological analysis was conducted to identify hub genes using the CytoHubba plugin of Cytoscape. The study includes calculating their degree centrality, closeness centrality, betweenness centrality, and maximum neighborhood component (MNC). For each metric, the top 20 hub genes were identified and selected. Then, the hub genes common to all four metrics were identified and used in the study. A separate PPI network was also generated to visualize the connection of these hub genes.

2.6. Survival Analysis of Hub Genes

Survival analysis, including overall survival (OS) and disease-free survival (DFS), was conducted using the Gene Expression Profiling Interactive Analysis (GEPIA) (http://gepia.cancer-pku.cn/, accessed on 6 September 2024) webserver for the identified hub genes. The KIRC patients were categorized into high- and low-expression groups based on the median expression levels of the cancer samples. Then, a plot was generated using the Kaplan–Meier method. Hub genes with a p-value less than 5 were considered statistically significant and included in the study.

2.7. Drug Repurposing Analysis

The top hub genes in each module were classified as upregulated or downregulated using GEO2R. These hub genes were used for drug repurposing analysis using the Drug Repurposing Encyclopedia (DRE) (https://www.drugrep.org/drugrepurposinganalysis, accessed on 6 September 2024) webserver. These identify potential drugs based on transcription profiles from MSigDB (Molecular Signatures Database) and CMap (Connectivity Map) [28]. Only the drugs with a false discovery rate (FDR) below 0.05, a negative Tau, and a known mechanism of action were considered in the analysis.

3. Results

3.1. Weighted Gene Co-Expression Network Analysis (WGCNA)

3.1.1. Scale-Free Network Analysis of the ccRCC Datasets

In WGCNA, gene expression matrices are normalized and outliers are removed from the data [21,27]. This results in filtered data (Figure S1) with outliers removed (Figure S2). Following this, 26,434 common genes are used to build the networks. The large number of genes in the analysis suggests robust analysis [23,24,26,29]. Figure 1 presents the scale-free topology fit index as a function of soft threshold (β) values (1–20). From this figure, the point at which the fit stabilized was at β = 10. Thus, β = 10 was selected to analyze all the datasets since this may promote a robust gene network with biologically relevant cluster construction.
Figure 2 illustrates that the log–log plot for the ccRCC S1 dataset had the highest fit, with an R2 value of 0.96. This indicates that the S1 dataset is the most appropriate for constructing the WGCNA network and identifying modules due to its relevant biological expression patterns [30]. Along with that, an R2 value near 0.9 is also considered ideal. The higher R2 value supports the idea that biological networks tend to exhibit scale-free characteristics, making this dataset a strong candidate for representing functionally important networks.

3.1.2. Network Construction and Module Identification

In WGCNA, meta-analysis consists of mapping module eigengenes from different datasets onto a reference dataset. This influences the robustness of the network and clustering resolution based on TOM 23. For this study, the ccRCC S1 dataset was selected as the reference. It also represents the early stage of ccRCC. A soft threshold power of β = 10 was used to construct the adjacency matrix. The S1 dataset was selected as the reference for the analysis, and the identified modules are displayed in Figure 3. This dataset’s expression profiles may provide a basis for understanding ccRCC S1 to S4. The sensitivity plots (Figure S3) also suggested that these module sets were stable even as sensitivity increased, presumably because of either strong gene correlations or fewer significant clusters. WGCNA identified seven gene co-expression modules as follows, each with a distinct color and a total number of genes: blue (2500 genes), brown (2497 genes), red (1242 genes), grey (2500 genes), green (1557 genes), yellow (2320 genes), and turquoise (2500 genes).

3.2. Module Preservation Analysis

Module preservation examines how well-defined modules are conserved across ccRCC datasets. This analysis reflects the reproducibility of co-expression networks and biological relevance. Module preservation in the ccRCC S1 dataset was assessed from the S2–S4 datasets. Modules with a Z-score value greater than 10 were considered highly preserved, and larger modules represented stronger patterns of connectivity [23,24]. Figure 4 illustrates the Z-score values of each module’s genes from different stages. Seven modules were generated with high preservations of blue, brown, and red. Although the turquoise module had 2500 genes, it was not strongly consistent across all datasets and was excluded. The blue module showed the highest score value in all stages. The number of modules passing the threshold was almost equal across datasets, except for S3, which had a higher score value than S2, before somewhat decreasing in S4. This is perhaps because S4 included genes that were not expressed during earlier stages of ccRCC.

3.3. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathways Analyses

The datasets were analyzed using eigengene-based connectivity (kME) by correlating the expression profile of each gene with module eigengenes. The genes were ranked by their kME values. The top genes were selected for functional analysis using DAVID GO categories (BP, CC, MF) and KEGG pathways. Enriched GO and KEGG terms for each module are provided in Table A1, Table A2, Table A3, Table A4 and Table A5, and the top terms are presented in Figure 5. Blue, red, and brown modules are mainly involved in transcription regulation. These modules are active in the cytosol, plasma membrane, and cytoplasm and are all associated with protein binding. KEGG pathway analysis indicated that these modules participate in pathways related to ccRCC through inflammatory pathways.

3.4. Protein–Protein Interaction (PPI) Networks and Hub Genes Analysis

PPIs of the identified genes were obtained from the STRING database. STRING was used to construct PPI networks for each preserved module with a high-confidence score (>0.7). The top 20 hub genes in each module were determined using degree centrality, closeness centrality, betweenness centrality, and MNC topology metrics. Figure 6 shows these hub genes, with red having the highest interaction scores. The high score of interaction reveals that these hub genes have a very significant association with ccRCC. Moreover, the genes overlapped between these metrics were defined as final hub genes (Figure 7) and used for further analysis.

3.5. Survival Analysis

All the hub genes were screened for survival analysis using GEPIA, but only six genes yielded statistically significant results. Their Kaplan–Meier survival plots are presented in Figure 8. MAPK8, BCL2, SMAD3, MAPK1, CREBBP, and EP300 are associated with much better survival rates since their analysis showed lower chances of death and recurrence of the disease. On the other hand, SOX2 is associated with poor OS, and its role in disease recurrence is less certain. Collectively, these findings indicate that these genes have the potential to be biomarkers in ccRCC.

3.6. Drug Repurposing Analysis

GEO2R was used to classify the top hub genes as upregulated or downregulated genes based on the positive and negative values of fold-change (FC). The DRE webserver analyzed these gene profiles to identify promising drug candidates. Lower false discovery rates (FDR) and more negative Tau values provide more reliable results and stronger effects on the target genes [24]. The top drug candidates identified for upregulated hub genes are ziprasidone, etazolate, trequinsin, dicycloverine, and decitabine. On the other hand, the leading candidates of drugs in downregulated hub genes include fentiazac, asenapine, KU-60019, pazopanib, and niacin. Detailed information about these drugs is included in Table 2.

4. Discussion

4.1. Gene Expression Modules and Pathway Analysis across the Datasets

ccRCC is the most common type of renal cancer. It progresses rapidly and often metastasizes to other parts of the body. This makes the prognosis difficult to determine [31,32]. Most cases of ccRCC are generally diagnosed in their later stages, so standard treatments are less effective; this is why new diagnostic methods are needed and being developed [32,33]. To address these problems, the present study carried out WGCNA analysis on samples of ccRCC from S1 to S4. This research used the S1 dataset to study the molecular and genetic characteristics of ccRCC and identify the key genes and pathways present from S1 to S4. Following this, the identified targets were analyzed to know if they may serve as potential biomarkers or targets for ccRCC. The KEGG pathway analysis of highly preserved gene modules (Table A1) suggests that these modules are linked to inflammatory pathways that may drive tumor development (Figure 9).
The analysis of S1 and S4 datasets revealed that specific pathways are present at certain stages of ccRCC (Figure 10). In S1, the HIF-1, TGF-β, and PI3K-Akt pathways suggest early tumor adaptation. HIF-1, mediated by the oxygen-sensitive HIF-1α and HIF-1β, helps cancer cells adapt to low oxygen. It is also linked with inflammation since it can regulate inflammatory components [34,35], thus promoting tumor development in ccRCC [36,37]. Enhanced TGF-β signaling can foster a proinflammatory microenvironment [14,38,39,40], reducing the effectiveness of anti-cancer therapies [41]. The PI3KT-AkT pathway has been known to stimulate inflammation processes and interact with other inflammatory pathways [42], which also accelerates tumor progression [43,44,45,46] and chemotherapy resistance [47]. The Ras and MAPK pathways were found to be present in S2 tumors, which may indicate heightened aggression and immune response in ccRCC [35,36,37,38]. Dysregulated RAS promotes inflammatory cytokine secretion [48,49], influencing ccRCC. The dysregulated MAPK pathway in ccRCC [43,50] leads to the release of cytokines such as IL-6, IL-8, and TNF-α [44,45,50].
Hippo signaling was detected in S3. Hippo signaling shows an inverse correlation with the proliferation and invasion of tumors [46,51,52]. Abnormal Hippo can increase cytokine production and activate other cancer pathways like JAK-STAT [47,51,53]. In S4, the NF-κB pathway and viral carcinogenesis are present. The NF-κB activated by IL-1 and TNFα connects inflammation with tumorigenesis [54,55,56]. NF-κB drives HIF-1α [57,58]. Hippo, MAPK, PI3K-AKT, and NF-κB pathways interact with each other, supporting inflammatory signals, cancer cell growth, and therapy resistance [44,47,59,60]. Additionally, viral infections like HPV and CMV associated with inflammation may significantly affect cancer treatment and growth [42,61,62,63]. The findings suggest that these pathways are involved in ccRCC S1–S4, indicating their role as drivers of ccRCC development.
The S1 dataset as a reference set helps track genes and pathways that may be present from S1 to S4. This may facilitate the identification of potential targets associated with ccRCC. The blue, brown, and red co-expression modules were preserved across datasets. Functional annotation linked these modules to inflammation-driven cancer pathways (Table A3, Table A4 and Table A5. Studying pathways present at different/certain stages of ccRCC may provide additional knowledge regarding ccRCC, which can help identify potential targets for specific stages of the disease.

4.2. Module Key Hub Genes and Their Protein Functions

4.2.1. Involvement of Hub Genes in Inflammation-Related Pathways Associated with ccRCC

The identified hub genes were found to be connected in these pathways. KEGG pathway analysis confirmed the involvement of hub genes in RCC, since RCC was obtained in the analysis (Table A1). Upregulated genes EGFR, CD44, and SOX2 in the red module are linked with the HIF-1 signaling pathway. EGFR, overexpressed in ccRCC and activated by IL-6 and TNF-α, is less effective as a target due to the lack of mutations or gene amplification [64,65]. Its membranous expression in RCC tumors [66], compared to its cytoplasmic location in normal kidney tissue, is associated with poorer prognosis and advanced stages [67]. HIF-1 also drives CD44 [68,69] upregulation in ccRCC [70,71,72] and correlates with poor survival and higher tumor grade [73,74]. SOX2, upregulated by HIF-1, enhances pro-inflammatory signaling and cancer stem cell-like properties, contributing to therapy resistance [75,76,77] with poorer overall survival (Figure 8E). In ccRCC, the NF-κB pathway mediates cytokines and chemokines [55,78,79,80,81,82]. Loss of VHL amplifies NF-κB activation, thus upregulating BCL2 [14,79,80]. BCL2 upregulated in the brown and red modules showed that it is associated with better survival and reduced risk of death and recurrence in kidney cancer (Figure 8A), establishing its role as potential marker. Venetoclax, a BCL2 inhibitor, was found to be effective in cancers such as leukemia, but continuous use of this agent can lead to drug resistance [83,84]. ATM enhances NF-κB, and its expression level could serve as a biomarker for ccRCC [65,85]. ATM is also involved in resistance to radio- and chemo-therapeutic treatments [65].
Upregulated SRC was found in the brown and red modules. Studies suggest that targeting SRC may disrupt pathways in ccRCC [66,86]. Chemokine can also activate the RAS signaling pathway. Cytokine-based therapies and immune checkpoint inhibitors are being developed to target chemokine signaling to address cancer cells immunity and address their resistance [87,88,89,90,91]. The Hippo pathway, linked with RCC [52], involves SMAD3 [53,92]. SMAD3 activates inflammation and interacts with Hippo effector YAP/TAZ [14,93]. SMAD3 also showed good survival analysis with reduced risk of death and recurrence (Figure 8D), thus targeting SMAD3 in Hippo may effectively inhibit ccRCC [47,59].
The MAPK pathway, often activated in cancer [94], upregulates MAPK1 and MAPK8 [95,96] found in the blue and red modules. The survival analysis also indicated that high levels of MAPK1 and MAPK8 are associated with better survival outcomes (Figure 8E,F). Research shows that downregulated MYPT1 in ccRCC inhibits MAPK8 activation, reducing metastasis [97]. The PI3K-Akt pathway regulates genes such as the upregulated GSK3B found in the red and brown modules. This pathway is frequently dysregulated in RCC since it interacts with the VHL/HIF [98,99,100], and a study found that targeting PI3K/Akt, along with its component, with inhibitors like LY294002 and wortmannin reduces inhibit RCC [101]. Overall, these pathways and genes are found to be associated with RCC [17,100].

4.2.2. Protein Chaperones and Epigenetic Regulators in Inflammation and Tumor Development in ccRCC

Molecular chaperones are known to be involved in protein folding but are now recognized in inflammation [102,103] and, under certain conditions, may contribute to tissue pathology [104,105]. Additionally, epigenetic regulators are also linked to inflammation [106,107,108]. Upregulated protein chaperones like HSP90AA1 were observed in the blue module, while epigenetic regulators such as HDAC1 and CREBBP were observed in the blue, brown, and red modules. HSP90AA1 maintains the stability of proteins related to signal transduction [109,110,111]. Epigenetic alterations in ccRCC influence inflammation through histones [107,112,113]. HDACs such as HDAC1 modulate the production of cytokines [107,112,114,115,116]. CREBBP is associated with inflammation [117]. Studies suggest that dysregulation of CREBBP, along with its partner EP300, drives the development of cancer and chemoresistance of several malignancies and affects tumor immune responses. Thus, this makes CREBBP/EP300 promising targets in cancer studies, as supported with the survival analysis (Figure 8B,C) [118]. Despite their potential as a target or biomarker, further research is still needed to validate their roles in ccRCC.

4.2.3. Metabolic Reprogramming in ccRCC

The constructed gene network suggests that it may be associated with the metabolic reprogramming of ccRCC supported by the PI3K/Akt and HIF-1 pathways. The yellow module (Table A2) highly represented the role of metabolic pathways. Several upregulated genes like INS, PRKACG, and CALML3 found in the brown and red modules were involved in these pathways. Metabolic reprogramming has been associated with tumor development and progression in ccRCC. The Warburg effect involves conditions of normal oxygen, resulting in the generation of lactate, along with an acidic microenvironment that boosts the migratory capabilities of the cancer cells. Glutamine metabolism is elevated in ccRC cells. As in most cases of ccRCC, mutation of the VHL gene disrupts the degradation of HIFs, initiating metabolic changes [119,120].
NMR studies have identified metabolites like lactate and glutamate in RCC [121]. In ccRCC, glutamate influences the PI3K/AKT pathway, with glutamate derived from glutamine via glutaminase [121,122]. The downregulation of GLUD1, which converts glutamate to α-ketoglutarate, is linked to increased malignancy and poor prognosis by enhancing PI3K/Akt/mTOR. Thus, GLUD1 is a potential target in ccRCC [122,123,124,125]. Another study also identified increased levels of creatine, alanine, lactate, and pyruvate using 1H-NMR. Metabolites findings with transcriptomic data from ccRCC tumor stem cells showed changes with HIF-α. Pyruvate, betaine, and creatinine were also found in HIF-1 in ccRCC. Pyruvate is an intermediate in glycolysis and thereby reflects increased glycolytic activity by HIF-1α through the upregulation of genes related to glycolysis. Betaine intersects with HIF-1α involved in methionine and choline metabolism. Creatinine, known for renal function and dysfunction, is indirectly related to HIF-1α activity [126]. Moreover, lactate stabilizes HIF-1α, making it a potential indicator of the HIF-1α pathway in ccRCC [34,126].

4.3. Viral Infections Associated with ccRCC

Viral infections significantly contribute to global cancer rates [63,127,128,129]. One study showed that the modules are linked to viral infections, including HPV and CMV [130]. These viral infections in ccRCC are known to cause inflammation [61,131,132]. CMV initiates inflammatory responses through CD14 and TLR2 [62]. CMV infection can be more severe in ccRCC patients compared to those with healthy immune systems, and cancer treatment increases the risk of infections like CMV, which can cause serious health issues [133]. The link between HPV and renal cancer is not fully understood yet. Some studies find no significant association, while others suggest HPV may contribute to RCC development or progression [131].

4.4. Drug Repurposing Based on Gene Signatures

New chemotherapeutic drugs have advanced tumor treatment but their development process is expensive and time consuming. Also, not all developed drugs are effective in clinical trials. Thus, repurposing existing drugs may offer a quicker and more economical solution, as these drugs have already undergone safety testing and are used to treat other diseases [134,135]. Among the upregulated hub genes, ziprasidone (ZIP) is the top candidate repurposed drug for ccRCC (Table 2). ZIP is an antipsychotic medication [136]. The effects of ZIP on inflammatory pathways like NF-κB and HIF-1 are not well established but studies found that it may inhibit several cancers such as pancreatic cancer [137] and breast cancer [138]. For instance, ZIP inhibits GOT1 to disrupt metabolic processes and reduce pancreatic cancer cell growth and has shown anti-cancer properties in xenograft models in preclinical studies [137]. Through in vitro studies, it was found that ZIP selectively inhibits aromatase with less toxicity than existing inhibitors in breast cancer [138].
Antipsychotic drugs such as haloperidol, chlorpromazine, thioridazine, and penfluridol are now being studied for their potential anti-cancer properties [134]. Thioridazine has also been studied in the PI3K/AKT and NF-κB pathways [134,139,140,141]. Thus, the findings suggest that ZIP could be repurposed as a potential treatment for ccRCC since it was already tested in other cancer treatments and may inhibit pathways where other antipsychotic drugs were already being studied.
For downregulated hub genes, fentiazac, a nonsteroidal anti-inflammatory drug (NSAID), was the top candidate drug that may be repurposed for ccRCC (Table 2). The connection of inflammation and cancer has led to interest in NSAIDs for cancer treatment. NSAIDs like aspirin, ibuprofen, diclofenac, celecoxib, tepoxalin, naproxen, and indomethacin have demonstrated anti-cancer properties, and some are already in clinical trials [142,143,144,145,146,147,148]. Fentiazac has not yet been documented for cancer treatment, particularly ccRCC, but it may be repurposed to target ccRCC by inhibiting COX-1 and COX-2 [149], like other NSAIDs studied for cancer drug repurposing. As a COX-1 inhibitor, it may also inhibit pathways associated with ccRCC, such as MAPK and NF-κB, targeting genes like EGFR, MAPK1, and BCL2 [150,151]. These considerations suggest that fentiazac could be a potential candidate for ccRCC treatment.
Other drug candidates associated with upregulated hub genes are etazolate, trequinsin, dicycloverine, and decitabine. On the other hand, the candidates for downregulated hub genes are asenapine, KU-60019, Fr-180204, and niacin (Table 2). Neuroactive drugs such as etazolate (phosphodiesterase inhibitor) and asenapine (dopamine and serotonin receptor antagonist) may target ccRCC by the MAPK, PI3K/Akt, and HIF-1α pathways [152,153,154]. Trequinsin, a phosphodiesterase inhibitor, and dicycloverine (phosphodiesterase inhibitor) commonly used for gastrointestinal cramps [155] may reduce side effects. However, their relationship with ccRCC and other cancers has not been documented yet. Decitabine (DNA methyltransferase inhibitor) induces G2/M cell cycle arrest in ccRCC cells by inhibiting the p38/NF-κB pathway [156]. EX-527 (a SIRT inhibitor) can be repurposed to impact sirtuin pathways [157,158] that are associated with ccRCC. KU-60019 (an ATM kinase inhibitor) may improve cancer treatment by inhibiting DNA damage response and reducing AKT phosphorylation. Its ability to radiosensitize cancer cells supports its potential [159]. Fr-180204 (a MAP kinase inhibitor) can disrupt MAPK pathways, while cefotiam, an antibiotic [160], and niacin, a vitamin B precursor [161], do not directly target ccRCC, but they may have indirect roles by affecting inflammation and metabolic processes related to ccRCC.
These potential drug candidates may help manage symptoms and inhibit ccRCC through several pathways. However, the results are based solely on theoretical data and hypotheses, highlighting a significant limitation of this study. Further experimental research, including wet lab experimentation, is necessary to validate these findings and determine their practical applicability. Nonetheless, our study provides a starting point for further investigation into correlating hub genes in ccRCC.

5. Conclusions

In the study, three significant highly preserved modules across different ccRCC stages obtained from GSE53757, GSE22541, GSE66272, and GSE73731 were identified. With WGCNA, the top hub genes were identified for each module and were used to determine functional annotation, pathways, and potential drug candidates that can be repurposed. The analysis provides insights about ccRCC through signaling pathways associated with inflammation. The top drug candidates identified for repurposing are ziprasidone (dopamine and serotonin receptor antagonist) and fentiazac (cyclooxygen-ase inhibitor). Other candidates are phosphodiesterase inhibitor, acetylcholine receptor antagonist, DNA methyltransferase inhibitor, an ATM kinase inhibitor, a MAP kinase inhibitor, and NAD precursors. These findings may inhibit ccRCC through the identified inflammatory pathways. While our study is limited by its in silico approach, the theoretical data provided can be assist in developing potential drug candidates or markers for ccRCC. The hub genes identified may also offer an understanding regarding the mech-anism of ccRCC. Moreover, further analysis is recommended such as using RNA-seq data to analyze ccRCC and compare it with DNA microarray. Additionally, experiments and wet lab research are recommended to confirm and apply these findings.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/app14198768/s1, Figure S1: Boxplot of ccRCC microarray datasets: (a) Stage 1, (b) Stage 2, (c) Stage 3, and (d) Stage 4 datasets; Figure S2. Sample clustering dendrogram for ccRCC: (a) Stage 1, (b) Stage 2, (c) Stage 3, and (d) Stage 4 datasets; Figure S3. TOM-based dissimilarity clustering of ccRCC (a) S1, (b) S2, (c) S3, and (d) S4 datasets.

Author Contributions

Conceptualization, K.S.S., M.A.O. and L.L.T.; methodology, K.S.S.; software, K.S.S.; validation, M.A.O.; formal analysis, K.S.S. and M.A.O.; investigation, K.S.S. and M.A.O.; resources, K.S.S. and L.L.T.; data curation, K.S.S. and M.A.O.; writing—original draft preparation, K.S.S.; writing—review and editing, K.S.S. and M.A.O.; visualization, K.S.S. and M.A.O.; supervision, L.L.T. and P.-W.T.; project administration, L.L.T. and P.-W.T.; funding acquisition, L.L.T. and P.-W.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The gene microarray datasets used for the study are openly available in the NCBI Gene Expression Omnibus (GEO) database under the accession IDs GSE53757, GSE22541, GSE66272, and GSE73731 datasets at https://www.ncbi.nlm.nih.gov/geo/, accessed on 12 June 2024.

Acknowledgments

The authors would like to extend their gratitude to Mapua University for their support. Along with that, we also would like to acknowledge several online platforms that helped us with our manuscript. Some figures were created with BioRender.com (accessed on 1 August 2024) and Canva.com (accessed on 1 August 2024). Along with that, grammatical checking was conducted using Grammarly.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Table A1. Top KEGG pathways analysis results in the highly preserved modules—Blue, Brown, and Red—from the Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.genome.jp/kegg (accessed on 28 June 2024)).
Table A1. Top KEGG pathways analysis results in the highly preserved modules—Blue, Brown, and Red—from the Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.genome.jp/kegg (accessed on 28 June 2024)).
KEGGTermCountp-Value
BluePathways in cancer681.27 × 10−5
Viral carcinogenesis335.01 × 10−5
Ras signaling pathway290.01091
TGF-β signaling pathway180.002582
Renal cell carcinoma161.63 × 10−4
BrownPathways in cancer652.69 × 10−6
PI3K-Akt signaling pathway463.74 × 10−5
MAPK signaling pathway374.46 × 10−4
Hippo signaling pathway237.59 × 10−4
Renal cell carcinoma110.017668
RedPathways in cancer662.69 × 10−6
MAPK signaling pathway431.50 × 10−6
PI3K-Akt signaling pathway415.81 × 10−4
Ras signaling pathway307.19 × 10−4
HIF-1 signaling pathway170.003199
Table A2. Top KEGG pathways analysis results in the other modules—Turquoise, Yellow, and Green—from the Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.genome.jp/kegg (accessed on 28 June 2024)).
Table A2. Top KEGG pathways analysis results in the other modules—Turquoise, Yellow, and Green—from the Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.genome.jp/kegg (accessed on 28 June 2024)).
KEGGTermCountp-Value
TurquoisePathways in cancer633.51 × 10−6
PI3K-Akt signaling pathway477.03 × 10−6
MAPK signaling pathway372.28 × 10−4
Hippo signaling pathway210.002751
mTOR signaling pathway190.013614
YellowMetabolic pathways1230.029441
Pathways in cancer606.99 × 10−5
EGFR tyrosine kinase inhibitor resistance142.31 × 10−3
NF-kB signaling pathway140.022381
HIF-1 signaling pathway140.029444
GreenPathways in cancer585.47 × 10−4
Human papillomavirus infection350.013714
Human cytomegalovirus infection308.63 × 10−4
Viral carcinogenesis240.014283
Viral myocarditis165.64 × 10−5
Table A3. Top GO and pathway analysis results for the blue module, obtained from the Gene Ontology database (http://www.geneontology.org, accessed on 28 June 2024).
Table A3. Top GO and pathway analysis results for the blue module, obtained from the Gene Ontology database (http://www.geneontology.org, accessed on 28 June 2024).
CategoryTermCountp-Value
BPGO:0045944~positive regulation of transcription by RNA polymerase II1522.35 × 10−14
GO:0045893~positive regulation of DNA-templated transcription944.34 × 10−11
GO:0006338~chromatin remodeling693.39 × 10−17
GO:0000398~mRNA splicing, via spliceosome472.93 × 10−14
GO:0008380~RNA splicing385.65 × 10−9
CCGO:0005829~cytosol6566.62 × 10−79
GO:0005634~nucleus6272.76 × 10−50
GO:0005737~cytoplasm5509.12 × 10−34
GO:0005654~nucleoplasm5442.40 × 10−85
GO:0000785~chromatin1201.89 × 10−8
MFGO:0005515~protein binding11167.48 × 10−69
GO:0003723~RNA binding2487.34 × 10−46
GO:0042802~identical protein binding1693.16 × 10−7
GO:0061629~RNA polymerase II-specific DNA-binding transcription factor binding314.44 × 10−6
GO:0140297~DNA-binding transcription factor binding303.22 × 10−6
Table A4. Top GO and pathway analysis results for the brown module, obtained from the Gene Ontology database (http://www.geneontology.org, accessed on 28 June 2024).
Table A4. Top GO and pathway analysis results for the brown module, obtained from the Gene Ontology database (http://www.geneontology.org, accessed on 28 June 2024).
CategoryTermCountp-Value
BPGO:0045944~positive regulation of transcription by RNA polymerase II1154.74 × 10−9
GO:0007165~signal transduction1144.41 × 10−9
GO:0000122~negative regulation of transcription by RNA polymerase II812.91 × 10−5
GO:0045893~positive regulation of DNA-templated transcription682.60 × 10−6
GO:0008380~RNA splicing385.65 × 10−9
CCGO:0005886~plasma membrane3895.98 × 10−14
GO:0005829~cytosol3632.61 × 10−8
GO:0005737~cytoplasm3456.44 × 10−5
GO:0005654~nucleoplasm2455.81 × 10−4
GO:0070062~extracellular exosome1881.76 × 10−12
MFGO:0005515~protein binding7908.86 × 10−14
GO:0042802~identical protein binding1332.09 × 10−5
GO:0004672~protein kinase activity273.93 × 10−3
GO:0005178~integrin binding221.55 × 10−4
GO:0004713~protein tyrosine kinase activity171.26 × 10−4
Table A5. Top GO and pathway analysis results for the red module, obtained from the Gene Ontology database (http://www.geneontology.org, accessed on 28 June 2024).
Table A5. Top GO and pathway analysis results for the red module, obtained from the Gene Ontology database (http://www.geneontology.org, accessed on 28 June 2024).
CategoryTermCountp-Value
BPGO:0045944~positive regulation of transcription by RNA polymerase II1187.69 × 10−11
GO:0007165~signal transduction1037.45 × 10−7
GO:0030154~cell differentiation583.70 × 10−5
GO:0045893~positive regulation of DNA-templated transcription560.0013
GO:0008284~positive regulation of cell population proliferation501.10 × 10−5
CCGO:0005737~cytoplasm3681.72 × 10−9
GO:0005829~cytosol3511.53 × 10−7
GO:0005654~nucleoplasm2405.72 × 10−4
GO:0048471~perinuclear region of cytoplasm546.57 × 10−3
GO:0032991~protein-containing complex498.41 × 10−3
MFGO:0005515~protein binding7474.49 × 10−8
GO:0042802~identical protein binding1294.40 × 10−5
GO:0106310~protein serine kinase activity341.76 × 10−3
GO:0045296~cadherin binding338.03 × 10−4
GO:0043565~sequence-specific DNA binding321.00 × 10−4

References

  1. Muglia, V.F.; Prando, A. Renal Cell Carcinoma: Histological Classification and Correlation with Imaging Findings. Radiol. Bras. 2015, 48, 166–174. [Google Scholar] [CrossRef] [PubMed]
  2. Kanwal, R. Metastasis in Renal Cell Carcinoma: Biology and Treatment. Adv. Cancer Biol.-Metastasis 2023, 7, 100094. [Google Scholar] [CrossRef]
  3. Sanchez, D.J.; Simon, M.C. Genetic and Metabolic Hallmarks of Clear Cell Renal Cell Carcinoma. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2018, 1870, 23–31. [Google Scholar] [CrossRef] [PubMed]
  4. Lam, J.; Klatte, T.; Breda, A. Staging of Renal Cell Carcinoma: Current Concepts. Indian J. Urol. 2009, 25, 446. [Google Scholar] [CrossRef] [PubMed]
  5. Cheaib, J.G.; Patel, H.D.; Johnson, M.H.; Gorin, M.A.; Haut, E.R.; Canner, J.K.; Allaf, M.E.; Pierorazio, P.M. Stage-Specific Conditional Survival in Renal Cell Carcinoma after Nephrectomy. Urol. Oncol. Semin. Orig. Investig. 2020, 38, 6.e1–6.e7. [Google Scholar] [CrossRef]
  6. Swami, U.; Nussenzveig, R.H.; Haaland, B.; Agarwal, N. Revisiting AJCC TNM Staging for Renal Cell Carcinoma: Quest for Improvement. Ann. Transl. Med. 2019, 7 (Suppl. S1), S18. [Google Scholar] [CrossRef]
  7. Kane, C.J.; Mallin, K.; Ritchey, J.; Cooperberg, M.R.; Carroll, P.R. Renal Cell Cancer Stage Migration. Cancer 2008, 113, 78–83. [Google Scholar] [CrossRef]
  8. Garfield, K.; LaGrange, C.A. Renal Cell Cancer; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  9. Warren, A.Y.; Harrison, D. WHO/ISUP Classification, Grading and Pathological Staging of Renal Cell Carcinoma: Standards and Controversies. World J. Urol. 2018, 36, 1913–1926. [Google Scholar] [CrossRef]
  10. Rathmell, W.K.; Chen, S. VHL Inactivation in Renal Cell Carcinoma: Implications for Diagnosis, Prognosis and Treatment. Expert. Rev. Anticancer Ther. 2008, 8, 63–73. [Google Scholar] [CrossRef]
  11. Seizinger, B.R.; Rouleau, G.A.; Ozelius, L.J.; Lane, A.H.; Farmer, G.E.; Lamiell, J.M.; Haines, J.; Yuen, J.W.M.; Collins, D.; Majoor-Krakauer, D.; et al. Von Hippel–Lindau Disease Maps to the Region of Chromosome 3 Associated with Renal Cell Carcinoma. Nature 1988, 332, 268–269. [Google Scholar] [CrossRef]
  12. Los, M.; Jansen, G.H.; Kaelin, W.G.; Lips, C.J.; Blijham, G.H.; Voest, E.E. Expression Pattern of the von Hippel-Lindau Protein in Human Tissues. Lab. Investig. 1996, 75, 231–238. [Google Scholar] [PubMed]
  13. Latif, F.; Tory, K.; Gnarra, J.; Yao, M.; Duh, F.-M.; Orcutt, M.L.; Stackhouse, T.; Kuzmin, I.; Modi, W.; Geil, L.; et al. Identification of the von Hippel-Lindau Disease Tumor Suppressor Gene. Science (1979) 1993, 260, 1317–1320. [Google Scholar] [CrossRef] [PubMed]
  14. Kruk, L.; Mamtimin, M.; Braun, A.; Anders, H.-J.; Andrassy, J.; Gudermann, T.; Mammadova-Bach, E. Inflammatory Networks in Renal Cell Carcinoma. Cancers 2023, 15, 2212. [Google Scholar] [CrossRef]
  15. Mertz, K.D.; Demichelis, F.; Kim, R.; Schraml, P.; Storz, M.; Diener, P.-A.; Moch, H.; Rubin, M.A. Automated Immunofluorescence Analysis Defines Microvessel Area as a Prognostic Parameter in Clear Cell Renal Cell Cancer. Hum. Pathol. 2007, 38, 1454–1462. [Google Scholar] [CrossRef]
  16. Qian, C.; Huang, D.; Wondergem, B.; Teh, B.T. Complexity of Tumor Vasculature in Clear Cell Renal Cell Carcinoma. Cancer 2009, 115 (Suppl. S10), 2282–2289. [Google Scholar] [CrossRef]
  17. de Vivar Chevez, A.R.; Finke, J.; Bukowski, R. The Role of Inflammation in Kidney Cancer. In Inflammation and Cancer; Springer: Basel, Switzerland, 2014; pp. 197–234. [Google Scholar] [CrossRef]
  18. Godwin, J.L.; Zibelman, M.; Plimack, E.R.; Geynisman, D.M. Immune Checkpoint Blockade as a Novel Immunotherapeutic Strategy for Renal Cell Carcinoma: A Review of Clinical Trials. Discov. Med. 2014, 18, 341–350. [Google Scholar]
  19. Hwang, A.; Mehra, V.; Chhetri, J.; Ali, S.; Tran, M.; Roddie, C. Current Treatment Options for Renal Cell Carcinoma: Focus on Cell-Based Immunotherapy. Cancers 2024, 16, 1209. [Google Scholar] [CrossRef]
  20. Yang, J.; Wang, K.; Yang, Z. Treatment Strategies for Clear Cell Renal Cell Carcinoma: Past, Present and Future. Front. Oncol. 2023, 13, 1133832. [Google Scholar] [CrossRef] [PubMed]
  21. Randrup Hansen, C.; Grimm, D.; Bauer, J.; Wehland, M.; Magnusson, N. Effects and Side Effects of Using Sorafenib and Sunitinib in the Treatment of Metastatic Renal Cell Carcinoma. Int. J. Mol. Sci. 2017, 18, 461. [Google Scholar] [CrossRef]
  22. Jin, J.; Xie, Y.; Zhang, J.-S.; Wang, J.-Q.; Dai, S.-J.; He, W.; Li, S.-Y.; Ashby, C.R.; Chen, Z.-S.; He, Q. Sunitinib Resistance in Renal Cell Carcinoma: From Molecular Mechanisms to Predictive Biomarkers. Drug Resist. Updates 2023, 67, 100929. [Google Scholar] [CrossRef]
  23. Langfelder, P.; Horvath, S. WGCNA: An R Package for Weighted Correlation Network Analysis. BMC Bioinform. 2008, 9, 559. [Google Scholar] [CrossRef] [PubMed]
  24. Manuel, M.T.A.; Tayo, L.L. Navigating the Gene Co-Expression Network and Drug Repurposing Opportunities for Brain Disorders Associated with Neurocognitive Impairment. Brain Sci. 2023, 13, 1564. [Google Scholar] [CrossRef] [PubMed]
  25. Pasamba, E.C.; Orda, M.A.; Villanueva, B.H.A.; Tsai, P.-W.; Tayo, L.L. Transcriptomic Analysis of Hub Genes Reveals Associated Inflammatory Pathways in Estrogen-Dependent Gynecological Diseases. Biology 2024, 13, 397. [Google Scholar] [CrossRef] [PubMed]
  26. Orda, M.A.; Fowler, P.M.P.T.; Tayo, L.L. Modular Hub Genes in DNA Microarray Suggest Potential Signaling Pathway Interconnectivity in Various Glioma Grades. Biology 2024, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  27. Lv, J.-H.; Hou, A.-J.; Zhang, S.-H.; Dong, J.-J.; Kuang, H.-X.; Yang, L.; Jiang, H. WGCNA Combined with Machine Learning to Find Potential Biomarkers of Liver Cancer. Medicine 2023, 102, e36536. [Google Scholar] [CrossRef]
  28. Yin, X.; Wang, P.; Yang, T.; Li, G.; Teng, X.; Huang, W.; Yu, H. Identification of Key Modules and Genes Associated with Breast Cancer Prognosis Using WGCNA and CeRNA Network Analysis. Aging 2021, 13, 2519–2538. [Google Scholar] [CrossRef]
  29. Zhu, Y.; Yang, X.; Zu, Y. Integrated Analysis of WGCNA and Machine Learning Identified Diagnostic Biomarkers in Dilated Cardiomyopathy with Heart Failure. Front. Cell Dev. Biol. 2022, 10, 1089915. [Google Scholar] [CrossRef]
  30. Mailem, R.C.; Tayo, L.L. Drug Repurposing Using Gene Co-Expression and Module Preservation Analysis in Acute Respiratory Distress Syndrome (ARDS), Systemic Inflammatory Response Syndrome (SIRS), Sepsis, and COVID-19. Biology 2022, 11, 1827. [Google Scholar] [CrossRef]
  31. Makino, T.; Kadomoto, S.; Izumi, K.; Mizokami, A. Epidemiology and Prevention of Renal Cell Carcinoma. Cancers 2022, 14, 4059. [Google Scholar] [CrossRef]
  32. Schiavoni, V.; Campagna, R.; Pozzi, V.; Cecati, M.; Milanese, G.; Sartini, D.; Salvolini, E.; Galosi, A.B.; Emanuelli, M. Recent Advances in the Management of Clear Cell Renal Cell Carcinoma: Novel Biomarkers and Targeted Therapies. Cancers 2023, 15, 3207. [Google Scholar] [CrossRef]
  33. Mattila, K.E.; Vainio, P.; Jaakkola, P.M. Prognostic Factors for Localized Clear Cell Renal Cell Carcinoma and Their Application in Adjuvant Therapy. Cancers 2022, 14, 239. [Google Scholar] [CrossRef]
  34. Infantino, V.; Santarsiero, A.; Convertini, P.; Todisco, S.; Iacobazzi, V. Cancer Cell Metabolism in Hypoxia: Role of HIF-1 as Key Regulator and Therapeutic Target. Int. J. Mol. Sci. 2021, 22, 5703. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, Y.; Xing, C.; Deng, Y.; Ye, C.; Peng, H. HIF-1α Signaling: Essential Roles in Tumorigenesis and Implications in Targeted Therapies. Genes Dis. 2024, 11, 234–251. [Google Scholar] [CrossRef]
  36. Lombardi, O.; Robert Mole, D. HIF Pathways in Clear Cell Renal Cancer. In Biomarkers and Bioanalysis Overview; IntechOpen: London, UK, 2021. [Google Scholar] [CrossRef]
  37. Malkov, M.I.; Lee, C.T.; Taylor, C.T. Regulation of the Hypoxia-Inducible Factor (HIF) by Pro-Inflammatory Cytokines. Cells 2021, 10, 2340. [Google Scholar] [CrossRef] [PubMed]
  38. Reiss, M. TGF-β and Cancer. Microbes Infect. 1999, 1, 1327–1347. [Google Scholar] [CrossRef] [PubMed]
  39. Sanjabi, S.; Oh, S.A.; Li, M.O. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb. Perspect. Biol. 2017, 9, a022236. [Google Scholar] [CrossRef]
  40. Giarratana, A.O.; Prendergast, C.M.; Salvatore, M.M.; Capaccione, K.M. TGF-β Signaling: Critical Nexus of Fibrogenesis and Cancer. J. Transl. Med. 2024, 22, 594. [Google Scholar] [CrossRef] [PubMed]
  41. Derynck, R.; Turley, S.J.; Akhurst, R.J. TGFβ Biology in Cancer Progression and Immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 9–34. [Google Scholar] [CrossRef]
  42. Contreras, A.; Sánchez, S.A.; Rodríguez-Medina, C.; Botero, J.E. The Role and Impact of Viruses on Cancer Development. Periodontol. 2000 2024. [Google Scholar] [CrossRef]
  43. Guo, Y.; Pan, W.; Liu, S.; Shen, Z.; Xu, Y.; Hu, L. ERK/MAPK Signalling Pathway and Tumorigenesis (Review). Exp. Ther. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef]
  44. Jayakumar, T.; Lin, K.-C.; Chang, C.-C.; Hsia, C.-W.; Manubolu, M.; Huang, W.-C.; Sheu, J.-R.; Hsia, C.-H. Targeting MAPK/NF-κB Pathways in Anti-Inflammatory Potential of Rutaecarpine: Impact on Src/FAK-Mediated Macrophage Migration. Int. J. Mol. Sci. 2021, 23, 92. [Google Scholar] [CrossRef] [PubMed]
  45. Brägelmann, J.; Lorenz, C.; Borchmann, S.; Nishii, K.; Wegner, J.; Meder, L.; Ostendorp, J.; Ast, D.F.; Heimsoeth, A.; Nakasuka, T.; et al. MAPK-Pathway Inhibition Mediates Inflammatory Reprogramming and Sensitizes Tumors to Targeted Activation of Innate Immunity Sensor RIG-I. Nat. Commun. 2021, 12, 5505. [Google Scholar] [CrossRef]
  46. Lv, L.; Zhou, X. Targeting Hippo Signaling in Cancer: Novel Perspectives and Therapeutic Potential. MedComm 2023, 4, e375. [Google Scholar] [CrossRef] [PubMed]
  47. Yang, Y.; Zhou, H.; Huang, X.; Wu, C.; Zheng, K.; Deng, J.; Zheng, Y.; Wang, J.; Chi, X.; Ma, X.; et al. Innate Immune and Proinflammatory Signals Activate the Hippo Pathway via a Tak1-STRIPAK-Tao Axis. Nat. Commun. 2024, 15, 145. [Google Scholar] [CrossRef] [PubMed]
  48. Fernandez-Medarde, A.; Santos, E. Ras in Cancer and Developmental Diseases. Genes Cancer 2011, 2, 344–358. [Google Scholar] [CrossRef] [PubMed]
  49. Sadeghi Shaker, M.; Rokni, M.; Mahmoudi, M.; Farhadi, E. Ras Family Signaling Pathway in Immunopathogenesis of Inflammatory Rheumatic Diseases. Front. Immunol. 2023, 14, 1151246. [Google Scholar] [CrossRef]
  50. Xiao, K.; Liu, C.; Tu, Z.; Xu, Q.; Chen, S.; Zhang, Y.; Wang, X.; Zhang, J.; Hu, C.-A.A.; Liu, Y. Activation of the NF-κB and MAPK Signaling Pathways Contributes to the Inflammatory Responses, but Not Cell Injury, in IPEC-1 Cells Challenged with Hydrogen Peroxide. Oxid. Med. Cell. Longev. 2020, 2020, 1–14. [Google Scholar] [CrossRef]
  51. Guo, P.; Wang, Z.; Zhou, Z.; Tai, Y.; Zhang, A.; Wei, W.; Wang, Q. Immuno-Hippo: Research Progress of the Hippo Pathway in Autoimmune Disease. Immunol. Lett. 2021, 230, 11–20. [Google Scholar] [CrossRef]
  52. Yang, Y.; Gan, X.; Zhang, W.; Zhu, B.; Huangfu, Z.; Shi, X.; Wang, L. Research Progress of the Hippo Signaling Pathway in Renal Cell Carcinoma. Asian J. Urol. 2024. [Google Scholar] [CrossRef]
  53. Sun, Y.; Jin, D.; Zhang, Z.; Jin, D.; Xue, J.; Duan, L.; Zhang, Y.; Kang, X.; Lian, F. The Critical Role of the Hippo Signaling Pathway in Kidney Diseases. Front. Pharmacol. 2022, 13, 988175. [Google Scholar] [CrossRef]
  54. Pavitra, E.; Kancharla, J.; Gupta, V.K.; Prasad, K.; Sung, J.Y.; Kim, J.; Tej, M.B.; Choi, R.; Lee, J.-H.; Han, Y.-K.; et al. The Role of NF-κB in Breast Cancer Initiation, Growth, Metastasis, and Resistance to Chemotherapy. Biomed. Pharmacother. 2023, 163, 114822. [Google Scholar] [CrossRef] [PubMed]
  55. Lin, Y.; Bai, L.; Chen, W.; Xu, S. The NF-κB Activation Pathways, Emerging Molecular Targets for Cancer Prevention and Therapy. Expert. Opin. Ther. Targets 2010, 14, 45–55. [Google Scholar] [CrossRef]
  56. Guo, Q.; Jin, Y.; Chen, X.; Ye, X.; Shen, X.; Lin, M.; Zeng, C.; Zhou, T.; Zhang, J. NF-κB in Biology and Targeted Therapy: New Insights and Translational Implications. Signal Transduct. Target. Ther. 2024, 9, 53. [Google Scholar] [CrossRef]
  57. D’Ignazio, L.; Bandarra, D.; Rocha, S. NF-κB and HIF Crosstalk in Immune Responses. FEBS J. 2016, 283, 413–424. [Google Scholar] [CrossRef] [PubMed]
  58. D’Ignazio, L.; Batie, M.; Rocha, S. Hypoxia and Inflammation in Cancer, Focus on HIF and NF-κB. Biomedicines 2017, 5, 21. [Google Scholar] [CrossRef]
  59. Li, X.; Cho, Y.S.; Zhu, J.; Zhuo, S.; Jiang, J. The Hippo Pathway Effector YAP Inhibits HIF2 Signaling and CcRCC Tumor Growth. Cell Discov. 2022, 8, 103. [Google Scholar] [CrossRef] [PubMed]
  60. Morgos, D.-T.; Stefani, C.; Miricescu, D.; Greabu, M.; Stanciu, S.; Nica, S.; Stanescu-Spinu, I.-I.; Balan, D.G.; Balcangiu-Stroescu, A.-E.; Coculescu, E.-C.; et al. Targeting PI3K/AKT/MTOR and MAPK Signaling Pathways in Gastric Cancer. Int. J. Mol. Sci. 2024, 25, 1848. [Google Scholar] [CrossRef]
  61. Fernandes, J.V.; De Medeiros Fernandes, T.A.A.; De Azevedo, J.C.V.; Cobucci, R.N.O.; De Carvalho, M.G.F.; Andrade, V.S.; De Araújo, J.M.G. Link between Chronic Inflammation and Human Papillomavirus-Induced Carcinogenesis (Review). Oncol. Lett. 2015, 9, 1015–1026. [Google Scholar] [CrossRef]
  62. Compton, T.; Kurt-Jones, E.A.; Boehme, K.W.; Belko, J.; Latz, E.; Golenbock, D.T.; Finberg, R.W. Human Cytomegalovirus Activates Inflammatory Cytokine Responses via CD14 and Toll-Like Receptor 2. J. Virol. 2003, 77, 4588–4596. [Google Scholar] [CrossRef]
  63. Kellogg, C.; Kouznetsova, V.L.; Tsigelny, I.F. Implications of Viral Infection in Cancer Development. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2021, 1876, 188622. [Google Scholar] [CrossRef]
  64. Raji, L.; Tetteh, A.; Amin, A.R.M.R. Role of C-Src in Carcinogenesis and Drug Resistance. Cancers 2023, 16, 32. [Google Scholar] [CrossRef] [PubMed]
  65. Liu, Y.-P.; Zheng, C.-C.; Huang, Y.-N.; He, M.-L.; Xu, W.W.; Li, B. Molecular Mechanisms of Chemo- and Radiotherapy Resistance and the Potential Implications for Cancer Treatment. MedComm 2021, 2, 315–340. [Google Scholar] [CrossRef] [PubMed]
  66. Cossu-Rocca, P.; Muroni, M.R.; Sanges, F.; Sotgiu, G.; Asunis, A.; Tanca, L.; Onnis, D.; Pira, G.; Manca, A.; Dore, S.; et al. EGFR Kinase-Dependent and Kinase-Independent Roles in Clear Cell Renal Cell Carcinoma. Am. J. Cancer Res. 2016, 6, 71–83. [Google Scholar] [PubMed]
  67. Pu, Y.-S.; Huang, C.-Y.; Kuo, Y.-Z.; Kang, W.-Y.; Liu, G.-Y.; Huang, A.-M.; Yu, H.-J.; Lai, M.-K.; Huang, S.-P.; Wu, W.-J.; et al. Characterization of Membranous and Cytoplasmic EGFR Expression in Human Normal Renal Cortex and Renal Cell Carcinoma. J. Biomed. Sci. 2009, 16, 82. [Google Scholar] [CrossRef]
  68. Johnson, P.; Ruffell, B. CD44 and Its Role in Inflammation and Inflammatory Diseases. Inflamm. Allergy Drug Targets 2009, 8, 208–220. [Google Scholar] [CrossRef]
  69. Liang, G.; Li, S.; Du, W.; Ke, Q.; Cai, J.; Yang, J. Hypoxia Regulates CD44 Expression via Hypoxia-Inducible Factor-1α in Human Gastric Cancer Cells. Oncol. Lett. 2017, 13, 967–972. [Google Scholar] [CrossRef]
  70. Ryu, D.; Ryoo, I.; Kwak, M.-K. Overexpression of CD44 Standard Isoform Upregulates HIF-1α Signaling in Hypoxic Breast Cancer Cells. Biomol. Ther. 2018, 26, 487–493. [Google Scholar] [CrossRef]
  71. Papanastasiou, A.D.; Peroukidis, S.; Sirinian, C.; Arkoumani, E.; Chaniotis, D.; Zizi-Sermpetzoglou, A. CD44 Expression in Clear Cell Renal Cell Carcinoma (CcRCC) Correlates with Tumor Grade and Patient Survival and Is Affected by Gene Methylation. Genes 2024, 15, 537. [Google Scholar] [CrossRef]
  72. Kuwahara, G.; Hashimoto, T.; Tsuneki, M.; Yamamoto, K.; Assi, R.; Foster, T.R.; Hanisch, J.J.; Bai, H.; Hu, H.; Protack, C.D.; et al. CD44 Promotes Inflammation and Extracellular Matrix Production during Arteriovenous Fistula Maturation. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1147–1156. [Google Scholar] [CrossRef]
  73. Du, T.; Wu, Z.; Wu, Y.; Liu, Y.; Song, Y.; Ma, L. CD44 Is Associated with Poor Prognosis of CcRCC and Facilitates CcRCC Cell Migration and Invasion through HAS1/MMP9. Biomedicines 2023, 11, 2077. [Google Scholar] [CrossRef]
  74. Yaghobi, Z.; Movassaghpour, A.; Talebi, M.; Abdoli Shadbad, M.; Hajiasgharzadeh, K.; Pourvahdani, S.; Baradaran, B. The Role of CD44 in Cancer Chemoresistance: A Concise Review. Eur. J. Pharmacol. 2021, 903, 174147. [Google Scholar] [CrossRef] [PubMed]
  75. Njouendou, A.J.; Szarvas, T.; Tiofack, A.A.Z.; Kenfack, R.N.; Tonouo, P.D.; Ananga, S.N.; Bell, E.H.M.D.; Simo, G.; Hoheisel, J.D.; Siveke, J.T.; et al. SOX2 Dosage Sustains Tumor-Promoting Inflammation to Drive Disease Aggressiveness by Modulating the FOSL2/IL6 Axis. Mol. Cancer 2023, 22, 52. [Google Scholar] [CrossRef] [PubMed]
  76. Gu, W.; Wang, B.; Wan, F.; Wu, J.; Lu, X.; Wang, H.; Zhu, Y.; Zhang, H.; Shi, G.; Dai, B.; et al. SOX2 and SOX12 Are Predictive of Prognosis in Patients with Clear Cell Renal Cell Carcinoma. Oncol. Lett. 2018, 15, 4564–4570. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, S.; Xiong, X.; Sun, Y. Functional Characterization of SOX2 as an Anticancer Target. Signal Transduct. Target. Ther. 2020, 5, 135. [Google Scholar] [CrossRef]
  78. Castillo-Rodríguez, R.A.; Trejo-Solís, C.; Cabrera-Cano, A.; Gómez-Manzo, S.; Dávila-Borja, V.M. Hypoxia as a Modulator of Inflammation and Immune Response in Cancer. Cancers 2022, 14, 2291. [Google Scholar] [CrossRef]
  79. Deka, K.; Li, Y. Transcriptional Regulation during Aberrant Activation of NF-κB Signalling in Cancer. Cells 2023, 12, 788. [Google Scholar] [CrossRef]
  80. Karin, M. NF-κB as a Critical Link between Inflammation and Cancer. Cold Spring Harb. Perspect. Biol. 2009, 1, a000141. [Google Scholar] [CrossRef]
  81. Labrousse-Arias, D.; Martínez-Alonso, E.; Corral-Escariz, M.; Bienes-Martínez, R.; Berridy, J.; Serrano-Oviedo, L.; Conde, E.; García-Bermejo, M.-L.; Giménez-Bachs, J.M.; Salinas-Sánchez, A.S.; et al. VHL Promotes Immune Response against Renal Cell Carcinoma via NF-κB–Dependent Regulation of VCAM-1. J. Cell Biol. 2017, 216, 835–847. [Google Scholar] [CrossRef]
  82. Mirzaei, S.; Saghari, S.; Bassiri, F.; Raesi, R.; Zarrabi, A.; Hushmandi, K.; Sethi, G.; Tergaonkar, V. NF-κB as a Regulator of Cancer Metastasis and Therapy Response: A Focus on Epithelial–Mesenchymal Transition. J. Cell Physiol. 2022, 237, 2770–2795. [Google Scholar] [CrossRef]
  83. Campbell, K.J.; Tait, S.W.G. Targeting BCL-2 Regulated Apoptosis in Cancer. Open Biol. 2018, 8, 180002. [Google Scholar] [CrossRef]
  84. Kapoor, I.; Bodo, J.; Hill, B.T.; Hsi, E.D.; Almasan, A. Targeting BCL-2 in B-Cell Malignancies and Overcoming Therapeutic Resistance. Cell Death Dis. 2020, 11, 941. [Google Scholar] [CrossRef] [PubMed]
  85. Ren, W.; Xue, B.; Chen, M.; Liu, L.; Zu, X. Low Expression of ATM Indicates a Poor Prognosis in Clear Cell Renal Cell Carcinoma. Clin. Genitourin. Cancer 2019, 17, e433–e439. [Google Scholar] [CrossRef] [PubMed]
  86. Roelants, C.; Giacosa, S.; Pillet, C.; Bussat, R.; Champelovier, P.; Bastien, O.; Guyon, L.; Arnoux, V.; Cochet, C.; Filhol, O. Combined Inhibition of PI3K and Src Kinases Demonstrates Synergistic Therapeutic Efficacy in Clear-Cell Renal Carcinoma. Oncotarget 2018, 9, 30066–30078. [Google Scholar] [CrossRef] [PubMed]
  87. Slack, C. Ras Signaling in Aging and Metabolic Regulation. Nutr. Healthy Aging 2017, 4, 195–205. [Google Scholar] [CrossRef]
  88. Yi, M.; Li, T.; Niu, M.; Zhang, H.; Wu, Y.; Wu, K.; Dai, Z. Targeting Cytokine and Chemokine Signaling Pathways for Cancer Therapy. Signal Transduct. Target. Ther. 2024, 9, 176. [Google Scholar] [CrossRef]
  89. Ozga, A.J.; Chow, M.T.; Luster, A.D. Chemokines and the Immune Response to Cancer. Immunity 2021, 54, 859–874. [Google Scholar] [CrossRef]
  90. Li, H.; Wu, M.; Zhao, X. Role of Chemokine Systems in Cancer and Inflammatory Diseases. MedComm 2022, 3, e147. [Google Scholar] [CrossRef]
  91. Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the Cancer Microenvironment and Their Relevance in Cancer Immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef]
  92. Fu, M.; Hu, Y.; Lan, T.; Guan, K.-L.; Luo, T.; Luo, M. The Hippo Signalling Pathway and Its Implications in Human Health and Diseases. Signal Transduct. Target. Ther. 2022, 7, 376. [Google Scholar] [CrossRef]
  93. Hussein, N.A.; Ebid, S.A.; Ahmad, M.A.; Khedr, G.E.; Saad, D.M. The Possible Correlation between MiR-762, Hippo Signaling Pathway, TWIST1, and SMAD3 in Lung Cancer and Chronic Inflammatory Diseases. Sci. Rep. 2024, 14, 8246. [Google Scholar] [CrossRef]
  94. Cheng, Y.; Chen, J.; Shi, Y.; Fang, X.; Tang, Z. MAPK Signaling Pathway in Oral Squamous Cell Carcinoma: Biological Function and Targeted Therapy. Cancers 2022, 14, 4625. [Google Scholar] [CrossRef] [PubMed]
  95. Broom, O.J.; Widjaya, B.; Troelsen, J.; Olsen, J.; Nielsen, O.H. Mitogen Activated Protein Kinases: A Role in Inflammatory Bowel Disease? Clin. Exp. Immunol. 2009, 158, 272–280. [Google Scholar] [CrossRef] [PubMed]
  96. Manzoor, Z.; Koh, Y.-S. Mitogen-Activated Protein Kinases in Inflammation. J. Bacteriol. Virol. 2012, 42, 189. [Google Scholar] [CrossRef]
  97. Xie, Q.; Liu, R.; Zou, Z.; Feng, Y.; Huang, Y.; Xu, G.; Sun, W.; Liang, Y.; Zhong, W. MYPT1 Inhibits the Metastasis of Renal Clear Cell Carcinoma via the MAPK8/N-cadherin Pathway. FEBS Open Bio 2022, 12, 2083–2095. [Google Scholar] [CrossRef]
  98. Hoffmeister, L.; Diekmann, M.; Brand, K.; Huber, R. GSK3: A Kinase Balancing Promotion and Resolution of Inflammation. Cells 2020, 9, 820. [Google Scholar] [CrossRef]
  99. Guo, H.; German, P.; Bai, S.; Barnes, S.; Guo, W.; Qi, X.; Lou, H.; Liang, J.; Jonasch, E.; Mills, G.B.; et al. The PI3K/AKT Pathway and Renal Cell Carcinoma. J. Genet. Genom. 2015, 42, 343–353. [Google Scholar] [CrossRef]
  100. Banumathy, G.; Cairns, P. Signaling Pathways in Renal Cell Carcinoma. Cancer Biol. Ther. 2010, 10, 658–664. [Google Scholar] [CrossRef]
  101. Sourbier, C.; Lindner, V.; Lang, H.; Agouni, A.; Schordan, E.; Danilin, S.; Rothhut, S.; Jacqmin, D.; Helwig, J.-J.; Massfelder, T. The Phosphoinositide 3-Kinase/Akt Pathway: A New Target in Human Renal Cell Carcinoma Therapy. Cancer Res. 2006, 66, 5130–5142. [Google Scholar] [CrossRef]
  102. Henderson, B.; Pockley, A.G. Molecular Chaperones and Protein-Folding Catalysts as Intercellular Signaling Regulators in Immunity and Inflammation. J. Leukoc. Biol. 2010, 88, 445–462. [Google Scholar] [CrossRef]
  103. Zhao, H.; Raines, L.N.; Huang, S.C.-C. Molecular Chaperones: Molecular Assembly Line Brings Metabolism and Immunity in Shape. Metabolites 2020, 10, 394. [Google Scholar] [CrossRef]
  104. Neckers, L.; Tatu, U. Molecular Chaperones in Pathogen Virulence: Emerging New Targets for Therapy. Cell Host Microbe 2008, 4, 519–527. [Google Scholar] [CrossRef] [PubMed]
  105. Horváth, I.; Multhoff, G.; Sonnleitner, A.; Vígh, L. Membrane-Associated Stress Proteins: More than Simply Chaperones. Biochim. Biophys. Acta (BBA)-Biomembr. 2008, 1778, 1653–1664. [Google Scholar] [CrossRef] [PubMed]
  106. Vezzani, B.; Carinci, M.; Previati, M.; Giacovazzi, S.; Della Sala, M.; Gafà, R.; Lanza, G.; Wieckowski, M.R.; Pinton, P.; Giorgi, C. Epigenetic Regulation: A Link between Inflammation and Carcinogenesis. Cancers 2022, 14, 1221. [Google Scholar] [CrossRef] [PubMed]
  107. Angulo, J.C.; Manini, C.; López, J.I.; Pueyo, A.; Colás, B.; Ropero, S. The Role of Epigenetics in the Progression of Clear Cell Renal Cell Carcinoma and the Basis for Future Epigenetic Treatments. Cancers 2021, 13, 2071. [Google Scholar] [CrossRef]
  108. Stylianou, E. Epigenetics of Chronic Inflammatory Diseases. J. Inflamm. Res. 2018, 12, 1–14. [Google Scholar] [CrossRef]
  109. Wei, H.; Zhang, Y.; Jia, Y.; Chen, X.; Niu, T.; Chatterjee, A.; He, P.; Hou, G. Heat Shock Protein 90: Biological Functions, Diseases, and Therapeutic Targets. MedComm 2024, 5, e470. [Google Scholar] [CrossRef]
  110. Zuehlke, A.D.; Beebe, K.; Neckers, L.; Prince, T. Regulation and Function of the Human HSP90AA1 Gene. Gene 2015, 570, 8–16. [Google Scholar] [CrossRef]
  111. Ashburner, B.P.; Westerheide, S.D.; Baldwin, A.S. The P65 (RelA) Subunit of NF-κB Interacts with the Histone Deacetylase (HDAC) Corepressors HDAC1 and HDAC2 To Negatively Regulate Gene Expression. Mol. Cell Biol. 2001, 21, 7065–7077. [Google Scholar] [CrossRef]
  112. Yang, J.; Xu, J.; Wang, W.; Zhang, B.; Yu, X.; Shi, S. Epigenetic Regulation in the Tumor Microenvironment: Molecular Mechanisms and Therapeutic Targets. Signal Transduct. Target. Ther. 2023, 8, 210. [Google Scholar] [CrossRef]
  113. Hamilton, J.P. Epigenetics: Principles and Practice. Dig. Dis. 2011, 29, 130–135. [Google Scholar] [CrossRef]
  114. Bure, I.V.; Nemtsova, M.V.; Kuznetsova, E.B. Histone Modifications and Non-Coding RNAs: Mutual Epigenetic Regulation and Role in Pathogenesis. Int. J. Mol. Sci. 2022, 23, 5801. [Google Scholar] [CrossRef] [PubMed]
  115. Tang, J.; Yan, H.; Zhuang, S. Histone Deacetylases as Targets for Treatment of Multiple Diseases. Clin. Sci. 2013, 124, 651–662. [Google Scholar] [CrossRef]
  116. Husain, S.; Obert, E.; Singh, S.; Schnabolk, G. Inhibition of HDAC1 and 3 in the Presence of Systemic Inflammation Reduces Retinal Degeneration in a Model of Dry Age-Related Macular Degeneration. J. Ocul. Pharmacol. Ther. 2024, 40, 397–406. [Google Scholar] [CrossRef] [PubMed]
  117. Matt, T. Transcriptional Control of the Inflammatory Response: A Role for the CREB-Binding Protein (CBP). Acta Med. Austriaca 2002, 29, 77–79. [Google Scholar] [CrossRef]
  118. Zhu, Y.; Wang, Z.; Li, Y.; Peng, H.; Liu, J.; Zhang, J.; Xiao, X. The Role of CREBBP/EP300 and Its Therapeutic Implications in Hematological Malignancies. Cancers 2023, 15, 1219. [Google Scholar] [CrossRef]
  119. Zhu, H.; Wang, X.; Lu, S.; Ou, K. Metabolic Reprogramming of Clear Cell Renal Cell Carcinoma. Front. Endocrinol. 2023, 14, 1195500. [Google Scholar] [CrossRef]
  120. Wettersten, H.I.; Aboud, O.A.; Lara, P.N.; Weiss, R.H. Metabolic Reprogramming in Clear Cell Renal Cell Carcinoma. Nat. Rev. Nephrol. 2017, 13, 410–419. [Google Scholar] [CrossRef]
  121. Monteiro, M.S.; Barros, A.S.; Pinto, J.; Carvalho, M.; Pires-Luís, A.S.; Henrique, R.; Jerónimo, C.; Bastos, M.d.L.; Gil, A.M.; Guedes de Pinho, P. Nuclear Magnetic Resonance Metabolomics Reveals an Excretory Metabolic Signature of Renal Cell Carcinoma. Sci. Rep. 2016, 6, 37275. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, L.; Fang, Z.; Gao, P.; Zheng, J. GLUD1 Suppresses Renal Tumorigenesis and Development via Inhibiting PI3K/Akt/MTOR Pathway. Front. Oncol. 2022, 12, 975517. [Google Scholar] [CrossRef]
  123. Coloff, J.L.; Murphy, J.P.; Braun, C.R.; Harris, I.S.; Shelton, L.M.; Kami, K.; Gygi, S.P.; Selfors, L.M.; Brugge, J.S. Differential Glutamate Metabolism in Proliferating and Quiescent Mammary Epithelial Cells. Cell Metab. 2016, 23, 867–880. [Google Scholar] [CrossRef]
  124. Craze, M.L.; El-Ansari, R.; Aleskandarany, M.A.; Cheng, K.W.; Alfarsi, L.; Masisi, B.; Diez-Rodriguez, M.; Nolan, C.C.; Ellis, I.O.; Rakha, E.A.; et al. Glutamate Dehydrogenase (GLUD1) Expression in Breast Cancer. Breast Cancer Res. Treat. 2019, 174, 79–91. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, Y.-J.; Hu, Z.-L.; Hu, S.-D.; Li, Y.-X.; Xing, X.-W.; Yang, Y.; Du, X.-H. Glutamate Dehydrogenase Inhibits Tumor Growth in Gastric Cancer through the Notch Signaling Pathway. Cancer Biomark. 2019, 26, 303–312. [Google Scholar] [CrossRef] [PubMed]
  126. Ragone, R.; Sallustio, F.; Piccinonna, S.; Rutigliano, M.; Vanessa, G.; Palazzo, S.; Lucarelli, G.; Ditonno, P.; Battaglia, M.; Fanizzi, F.; et al. Renal Cell Carcinoma: A Study through NMR-Based Metabolomics Combined with Transcriptomics. Diseases 2016, 4, 7. [Google Scholar] [CrossRef]
  127. Bersanelli, M.; Casartelli, C.; Buti, S.; Porta, C. Renal Cell Carcinoma and Viral Infections: A Dangerous Relationship? World J. Nephrol. 2022, 11, 1–12. [Google Scholar] [CrossRef]
  128. Chaudhary, R.; Meher, A.; Krishnamoorthy, P.; Kumar, H. Interplay of Host and Viral Factors in Inflammatory Pathway Mediated Cytokine Storm during RNA Virus Infection. Curr. Res. Immunol. 2023, 4, 100062. [Google Scholar] [CrossRef]
  129. Sarier, M. Association between Human Papillomavirus and Urological Cancers: An Update. In Molecular Mechanisms in Cancer; IntechOpen: London, UK, 2022. [Google Scholar] [CrossRef]
  130. Read, S.A.; Douglas, M.W. Virus Induced Inflammation and Cancer Development. Cancer Lett. 2014, 345, 174–181. [Google Scholar] [CrossRef]
  131. Farhadi, A.; Behzad-Behbahani, A.; Geramizadeh, B.; Sekawi, Z.; Rahsaz, M.; Sharifzadeh, S. High-risk Human Papillomavirus Infection in Different Histological Subtypes of Renal Cell Carcinoma. J. Med. Virol. 2014, 86, 1134–1144. [Google Scholar] [CrossRef] [PubMed]
  132. Salehipoor, M.; Khezri, A.; Behzad-Behbahani, A.; Geramizadeh, B.; Rahsaz, M.; Aghdaei, M.; Afrasiabi, M.A. Role of Viruses in Renal Cell Carcinoma. Saudi J. Kidney Dis. Transpl. 2012, 23, 53–57. [Google Scholar]
  133. Rinaldi, I.; Muthalib, A.; Sutandar, J.W.; Kuncoro, H.A.; Harsono, B.I.; Susanto, N.; Setiawan, T.; Winston, K.; Dewantara, I.R.; Amin, I.F.; et al. Cytomegalovirus Infection in Patient with Clear Cell Renal Cell Carcinoma. Case Rep. Med. 2023, 2023, 1–6. [Google Scholar] [CrossRef]
  134. Vlachos, N.; Lampros, M.; Voulgaris, S.; Alexiou, G.A. Repurposing Antipsychotics for Cancer Treatment. Biomedicines 2021, 9, 1785. [Google Scholar] [CrossRef]
  135. Xia, Y.; Sun, M.; Huang, H.; Jin, W.-L. Drug Repurposing for Cancer Therapy. Signal Transduct. Target. Ther. 2024, 9, 92. [Google Scholar] [CrossRef] [PubMed]
  136. Duarte, T.; Barbisan, F.; do Prado-Lima, P.A.S.; Azzolin, V.F.; da Cruz Jung, I.E.; Duarte, M.M.M.F.; Teixeira, C.F.; Mastella, M.H.; da Cruz, I.B.M. Ziprasidone, a Second-Generation Antipsychotic Drug, Triggers a Macrophage Inflammatory Response in Vitro. Cytokine 2018, 106, 101–107. [Google Scholar] [CrossRef] [PubMed]
  137. Yang, Y.; Zheng, M.; Han, F.; Shang, L.; Li, M.; Gu, X.; Li, H.; Chen, L. Ziprasidone Suppresses Pancreatic Adenocarcinoma Cell Proliferation by Targeting GOT1 to Trigger Glutamine Metabolism Reprogramming. J. Mol. Med. 2022, 100, 599–612. [Google Scholar] [CrossRef] [PubMed]
  138. Sahu, A.; Ahmad, S.; Imtiyaz, K.; Kizhakkeppurath Kumaran, A.; Islam, M.; Raza, K.; Easwaran, M.; Kurukkan Kunnath, A.; Rizvi, M.A.; Verma, S. In-Silico and in-Vitro Study Reveals Ziprasidone as a Potential Aromatase Inhibitor against Breast Carcinoma. Sci. Rep. 2023, 13, 16545. [Google Scholar] [CrossRef] [PubMed]
  139. Cai, N.; Zhou, W.; Ye, L.-L.; Chen, J.; Liang, Q.-N.; Chang, G.; Chen, J.-J. The STAT3 Inhibitor Pimozide Impedes Cell Proliferation and Induces ROS Generation in Human Osteosarcoma by Suppressing Catalase Expression. Am. J. Transl. Res. 2017, 9, 3853–3866. [Google Scholar]
  140. Rho, S.B.; Kim, B.-R.; Kang, S. A Gene Signature-Based Approach Identifies Thioridazine as an Inhibitor of Phosphatidylinositol-3′-Kinase (PI3K)/AKT Pathway in Ovarian Cancer Cells. Gynecol. Oncol. 2011, 120, 121–127. [Google Scholar] [CrossRef]
  141. Kang, S.; Dong, S.M.; Kim, B.-R.; Park, M.S.; Trink, B.; Byun, H.-J.; Rho, S.B. Thioridazine Induces Apoptosis by Targeting the PI3K/Akt/MTOR Pathway in Cervical and Endometrial Cancer Cells. Apoptosis 2012, 17, 989–997. [Google Scholar] [CrossRef]
  142. Sousa, S.M.; Xavier, C.P.R.; Vasconcelos, M.H.; Palmeira, A. Repurposing Some of the Well-Known Non-Steroid Anti-Inflammatory Drugs (NSAIDs) for Cancer Treatment. Curr. Top. Med. Chem. 2023, 23, 1171–1195. [Google Scholar] [CrossRef]
  143. Schwandt, A.; Garcia, J.A.; Elson, P.; Wyckhouse, J.; Finke, J.H.; Ireland, J.; Triozzi, P.; Zhou, M.; Dreicer, R.; Rini, B.I. Clinical and Immunomodulatory Effects of Celecoxib Plus Interferon-Alpha in Metastatic Renal Cell Carcinoma Patients with COX-2 Tumor Immunostaining. J. Clin. Immunol. 2011, 31, 690–698. [Google Scholar] [CrossRef]
  144. Neve, V.; Kamble, A.; Karwa, P. Repurposing of Drug Aspirin in Colon Cancer: Therapeutic Approach. Clin. Cancer Investig. J. 2024, 13, 23–28. [Google Scholar] [CrossRef]
  145. Endo, H.; Yano, M.; Okumura, Y.; Kido, H. Ibuprofen Enhances the Anticancer Activity of Cisplatin in Lung Cancer Cells by Inhibiting the Heat Shock Protein 70. Cell Death Dis. 2014, 5, e1027. [Google Scholar] [CrossRef] [PubMed]
  146. McQuerry, J.A.; Chen, J.; Chang, J.T.; Bild, A.H. Tepoxalin Increases Chemotherapy Efficacy in Drug-Resistant Breast Cancer Cells Overexpressing the Multidrug Transporter Gene ABCB1. Transl. Oncol. 2021, 14, 101181. [Google Scholar] [CrossRef] [PubMed]
  147. Deb, J.; Majumder, J.; Bhattacharyya, S.; Jana, S.S. A Novel Naproxen Derivative Capable of Displaying Anti-Cancer and Anti-Migratory Properties against Human Breast Cancer Cells. BMC Cancer 2014, 14, 567. [Google Scholar] [CrossRef]
  148. Emam Kassab, S. Indomethacin from Anti-Inflammatory to Anticancer Agent. In Medicinal Chemistry; IntechOpen: London, UK, 2019. [Google Scholar] [CrossRef]
  149. Famaey, J.P.; Gaci, O.; Ginsberg, F. Fentiazac in the Treatment of Osteoarthritis and Tendinitis. Curr. Med. Res. Opin. 1983, 8, 675–681. [Google Scholar] [CrossRef]
  150. Purssell, E. Cyclooxygenase Inhibitors Inhibit Antibody Response through Interference with MAPK/ERK Pathways and BLIMP-1 Inhibition. Med. Hypotheses 2014, 83, 372–377. [Google Scholar] [CrossRef]
  151. Abo-El Fetoh, M.E.; Abdel-Fattah, M.M.; Mohamed, W.R.; Ramadan, L.A.A.; Afify, H. Cyclooxygenase-2 Activates EGFR–ERK1/2 Pathway via PGE2-Mediated ADAM-17 Signaling in Testosterone-Induced Benign Prostatic Hyperplasia. Inflammopharmacology 2023, 31, 499–516. [Google Scholar] [CrossRef]
  152. Marcade, M.; Bourdin, J.; Loiseau, N.; Peillon, H.; Rayer, A.; Drouin, D.; Schweighoffer, F.; Désiré, L. Etazolate, a Neuroprotective Drug Linking GABA A Receptor Pharmacology to Amyloid Precursor Protein Processing. J. Neurochem. 2008, 106, 392–404. [Google Scholar] [CrossRef] [PubMed]
  153. Balaraman, R.; Gandhi, H. Asenapine, a New Sublingual Atypical Antipsychotic. J. Pharmacol. Pharmacother. 2010, 1, 60–61. [Google Scholar] [CrossRef]
  154. DRUGBANK. Asenapine: Uses, Interactions, Mechanism of Action|DrugBank Online. Available online: https://go.drugbank.com/drugs/DB06216 (accessed on 9 August 2024).
  155. Aronson, J.K. Dicycloverine. In Meyler’s Side Effects of Drugs; Elsevier: Amsterdam, The Netherlands, 2016; p. 950. [Google Scholar] [CrossRef]
  156. Shang, D.; Han, T.; Xu, X.; Liu, Y. Decitabine Induces G2/M Cell Cycle Arrest by Suppressing P38/NF-κB Signaling in Human Renal Clear Cell Carcinoma. Int. J. Clin. Exp. Pathol. 2015, 8, 11140–11148. [Google Scholar]
  157. Broussy, S.; Laaroussi, H.; Vidal, M. Biochemical Mechanism and Biological Effects of the Inhibition of Silent Information Regulator 1 (SIRT1) by EX-527 (SEN0014196 or Selisistat). J. Enzym. Inhib. Med. Chem. 2020, 35, 1124–1136. [Google Scholar] [CrossRef]
  158. Yang, Y.; Liu, Y.; Wang, Y.; Chao, Y.; Zhang, J.; Jia, Y.; Tie, J.; Hu, D. Regulation of SIRT1 and Its Roles in Inflammation. Front. Immunol. 2022, 13, 831168. [Google Scholar] [CrossRef] [PubMed]
  159. Giacosa, S.; Pillet, C.; Séraudie, I.; Guyon, L.; Wallez, Y.; Roelants, C.; Battail, C.; Evrard, B.; Chalmel, F.; Barette, C.; et al. Cooperative Blockade of CK2 and ATM Kinases Drives Apoptosis in VHL-Deficient Renal Carcinoma Cells through ROS Overproduction. Cancers 2021, 13, 576. [Google Scholar] [CrossRef] [PubMed]
  160. Lambert, H.; Chen, M.; Cabral, C. Antimicrobial Resistance, Inflammatory Responses: A Comparative Analysis of Pathogenicities, Knowledge Hybrids and the Semantics of Antibiotic Use. Palgrave Commun. 2019, 5, 85. [Google Scholar] [CrossRef]
  161. Rad, E.Y.; Saboori, S.; Tammam, J.; Thondre, P.S.; Coe, S. The Effect of Niacin on Inflammatory Markers and Adipokines: A Systematic Review and Meta-Analysis of Interventional Studies. Eur. J. Nutr. 2024, 63, 2011–2024. [Google Scholar] [CrossRef]
Figure 1. Model fit for scale-free topology based on average gene connections at different soft-thresholding powers.
Figure 1. Model fit for scale-free topology based on average gene connections at different soft-thresholding powers.
Applsci 14 08768 g001
Figure 2. Linear relationship plot of the ccRCC S1 dataset with the X-axis showing node degree (k) and the Y-axis showing the probability (p(k)) of that degree.
Figure 2. Linear relationship plot of the ccRCC S1 dataset with the X-axis showing node degree (k) and the Y-axis showing the probability (p(k)) of that degree.
Applsci 14 08768 g002
Figure 3. Dendrogram of gene clustering and module identification for the ccRCC s1 dataset. Colors denote the gene co-expression modules associated with the sections of the dendrogram above.
Figure 3. Dendrogram of gene clustering and module identification for the ccRCC s1 dataset. Colors denote the gene co-expression modules associated with the sections of the dendrogram above.
Applsci 14 08768 g003
Figure 4. Module preservation in the ccRCC S1 network compared to (A) S2, (B) S3, and (C) S4 is indicated by Zsummary values. A Zsummary > 10 indicates strong preservation values.
Figure 4. Module preservation in the ccRCC S1 network compared to (A) S2, (B) S3, and (C) S4 is indicated by Zsummary values. A Zsummary > 10 indicates strong preservation values.
Applsci 14 08768 g004
Figure 5. The bubble plot displays GO terms and KEGG pathways enriched in each preserved module, based on DAVID.
Figure 5. The bubble plot displays GO terms and KEGG pathways enriched in each preserved module, based on DAVID.
Applsci 14 08768 g005
Figure 6. Top 20 hub genes of 3 highly preserved modules (blue, brown, and red) processed using (A1A3) MNC, (B1B3) degree, (C1C3) closeness, and (D1D3) betweenness centrality algorithms of Cytoscape.
Figure 6. Top 20 hub genes of 3 highly preserved modules (blue, brown, and red) processed using (A1A3) MNC, (B1B3) degree, (C1C3) closeness, and (D1D3) betweenness centrality algorithms of Cytoscape.
Applsci 14 08768 g006
Figure 7. Protein–protein interaction networks of the overlapping hub genes in the 3 modules (A) blue, (B) brown, and (C) red.
Figure 7. Protein–protein interaction networks of the overlapping hub genes in the 3 modules (A) blue, (B) brown, and (C) red.
Applsci 14 08768 g007
Figure 8. Kaplan–Meier survival curves for overall survival (OS) and disease-free survival (DFS) of hub genes with p < 0.05 in KIRC patients using GEPIA: (A) BCL2, (B) EP300, (C) CREBBP, (D) SMAD3, (E) MAPK1, (F) MAPK8, and (G) SOX2.
Figure 8. Kaplan–Meier survival curves for overall survival (OS) and disease-free survival (DFS) of hub genes with p < 0.05 in KIRC patients using GEPIA: (A) BCL2, (B) EP300, (C) CREBBP, (D) SMAD3, (E) MAPK1, (F) MAPK8, and (G) SOX2.
Applsci 14 08768 g008
Figure 9. Signaling pathways associated with inflammation in ccRCC.
Figure 9. Signaling pathways associated with inflammation in ccRCC.
Applsci 14 08768 g009
Figure 10. KEGG pathways associated with ccRCC. Arrows indicate that the specific signaling pathways are present at a certain stage.
Figure 10. KEGG pathways associated with ccRCC. Arrows indicate that the specific signaling pathways are present at a certain stage.
Applsci 14 08768 g010
Table 1. Information of the GEO datasets used in the study.
Table 1. Information of the GEO datasets used in the study.
Accession No.GSE53757GSE22541GSE66272GSE73731
ConditionccRCC Stage 1 (S1)ccRCC Stage 2 (S2)ccRCC Stage 3 (S3)ccRCC Stage 4 (S4)
TypeExpression Profiling by Array
PlatformGPL570—HG-U133 Plus 2 Affymetrix Human Genome U133 Plus 2.0 Array
SourcePrimary Tumor Samples
No. of Samples24212744
Table 2. Top candidates of upregulated and downregulated hub genes.
Table 2. Top candidates of upregulated and downregulated hub genes.
GenesDrugMechanismTauFDR
UpregulatedATM, CDC4, GSK3B, ITGA2B, MAPK8, MED1, PPARG, RPL5, CD44, HNRNPC, BCL2, CALML3, EGFR, GRB2, HDAC1, HSP90AA1, INS, PRKACG, SOX2, SRC, SMAD3, MAPK1, and TGFB1ZiprasidoneDopamine receptor antagonist, Serotonin receptor antagonist−99.00390.00884
EtazolatePhosphodiesterase inhibitor−98.92470.00207
TrequinsinPhosphodiesterase inhibitor−97.56320.00557
DicycloverineAcetylcholine receptor antagonist−97.264670.00936
DecitabineDNA methyltransferase inhibitor−96.34480.00267
DownregulatedPTK2, BDNF, CREBBP, DLG4, EP300, H3C12, and ESR1FentiazacCyclooxygenase inhibitor−99.64970.001254
AsenapineDopamine receptor antagonist, serotonin receptor antagonist−99.24150.007037
Ku-60019ATM kinase inhibitor−99.19590.007115
Fr-180204MAP kinase inhibitor−98.23940.000190
NiacinNAD precursor, vitamin B−98.22110.002544
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Suratos, K.S.; Orda, M.A.; Tsai, P.-W.; Tayo, L.L. Signaling Pathways in Clear Cell Renal Cell Carcinoma and Candidate Drugs Unveiled through Transcriptomic Network Analysis of Hub Genes. Appl. Sci. 2024, 14, 8768. https://doi.org/10.3390/app14198768

AMA Style

Suratos KS, Orda MA, Tsai P-W, Tayo LL. Signaling Pathways in Clear Cell Renal Cell Carcinoma and Candidate Drugs Unveiled through Transcriptomic Network Analysis of Hub Genes. Applied Sciences. 2024; 14(19):8768. https://doi.org/10.3390/app14198768

Chicago/Turabian Style

Suratos, Khyle S., Marco A. Orda, Po-Wei Tsai, and Lemmuel L. Tayo. 2024. "Signaling Pathways in Clear Cell Renal Cell Carcinoma and Candidate Drugs Unveiled through Transcriptomic Network Analysis of Hub Genes" Applied Sciences 14, no. 19: 8768. https://doi.org/10.3390/app14198768

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop