In recent years, different investigations in the area of multi-center genomics research from gene to systems level and next-generation sequencing, clarified the various mechanisms specifically involved in tumor progression. This study used bioinformatics analysis to further investigate the genes involved in GC pathogenesis (25).
Ten hub genes were identified in this study, including FN1, CTGF, CXCL5, IL6, ELN, ADAMTS2, MMP2, TP53, WISP1 and THBS1. The relationship between the overexpression of these genes and gastric cancer is an area of active scientific investigation. These genes have been implicated in various molecular and cellular pathways relevant to cancer initiation, progression, and metastasis. Overexpression of FN1, a key component of the extracellular matrix, may promote tumor cell adhesion, migration, and invasion, facilitating the metastatic potential of cancer cells (26) and may have the same roles in GC. Sun et al. did the same bioinformatics analysis and reported FN1 as one of the key genes involved in GC pathogenesis which confirms our results(27). CTGF, a growth factor associated with fibrosis and tissue remodeling, has been linked to the pathogenesis of gastric cancer, potentially influencing the tumor microenvironment, angiogenesis, and cell proliferation(28). CXCL5, an inflammatory chemokine, The presence of immune cells and inflammation in the microenvironment can potentially contribute to the development of gastric cancer, promoting tumor growth and metastasis(29). IL6, a pro-inflammatory cytokine, can stimulate various signaling pathways, leading to cell survival, proliferation, and immune modulation in gastric cancer, potentially contributing to tumor progression. Up-regulation of ELN, which encodes the elastin protein, may affect tissue integrity, elasticity, and angiogenesis, potentially impacting the invasive behavior of gastric cancer cells (30). ADAMTS2, involved in extracellular matrix remodeling, might influence tumor cell invasion and angiogenesis in gastric cancer (31). MMP2, a matrix metalloproteinase, has implications in extracellular matrix degradation, which is essential for tumor invasion and metastasis in gastric cancer (32). The preservation of genomic stability depends greatly on the TP53 gene, which functions as a tumor suppressor. Mutations or dysregulation of TP53 are commonly associated with gastric cancer, contributing to uncontrolled cell growth and reduced DNA repair (33). WISP1, which regulates the Wnt signaling pathway, may have a part in encouraging cell growth and movement in gastric cancer, which could aid in the progression of the tumor (34).
THBS1, an adhesive glycoprotein, is involved in angiogenesis and cell-matrix interactions, and its overexpression may contribute to tumor angiogenesis and metastasis in gastric cancer. In conclusion(35), the overexpression or up-regulation of these genes is interconnected with various cellular processes and molecular pathways relevant to gastric cancer. However, the specific roles of each gene in the context of gastric cancer initiation, progression, and metastasis are complex and multifaceted. Further comprehensive research is essential to elucidate the precise contributions of these genes, potentially leading to the development of targeted therapeutic strategies and the identification of biomarkers for gastric cancer management.
The results of our study showed that the PI3K-Akt signaling pathway is the prominent pathway affected in GC. The PI3K-Akt pathway is commonly disrupted in GC, with multiple essential components showing alterations. Activation of this pathway leads to enhanced tumor growth, survival, metastasis, angiogenesis, and resistance to treatments in GC(36). The primary genomic changes causing abnormal PI3K-Akt activation involve PIK3CA amplifications/mutations, loss of PTEN, and the overexpression of proteins that activate chemical signaling pathways called receptor tyrosine kinases (such as EGFR, HER2, cMET). Clinical investigations have linked active PI3K-Akt signaling to aggressive disease, advanced tumor stages, metastasis, and unfavorable prognoses in patients with GC. Experimental studies indicate that inhibiting PI3K-Akt signaling curbs GC cancer cell proliferation induces apoptosis, and heightens sensitivity to chemotherapy and radiotherapy(37). Preliminary results from early-stage trials of PI3K/Akt/mTOR inhibitors, either alone or combined, have demonstrated potential efficacy against advanced GC. Noteworthy challenges encompass treatment-related toxicity, acquired resistance, and the subset of patients who respond positively to PI3K pathway inhibition, thus emphasizing the need to explore predictive biomarkers.
According to research, an increased amount of CTGF, FN1, IL-6, THBS1, and WISP1 genes has been found to be linked to shorter survival times in patients with GC. Further investigations are warranted to elucidate their potential as therapeutic targets. In the context of GC, CTGF overexpression augments tumor growth, invasion, and resistance to chemotherapy, partly through the activation of PI3K-Akt signaling. This interaction is facilitated by the direct binding of CTGF to the p85 subunit of PI3K. FN1 upregulation triggers FAK/Src signaling, which cross-communicates and amplifies PI3K-Akt signaling in GC cells, thereby promoting migration and invasion processes. Similarly, IL-6 overexpression in GC fuels PI3K-Akt signaling activation via the GP130/JAK/STAT3 cascade, resulting in a heightened advantage in terms of proliferation and survival. THBS1 upregulation in GC leads to PI3K-Akt signaling activation through integrin-mediated pathways, fostering increased tumor growth and resistance to therapy. WISP1, contributing to GC progression, partially activates the PI3K-Akt/mTOR axis, consequently intensifying proliferation, invasion, and angiogenesis. Importantly, a reciprocal relationship emerges, wherein PI3K-Akt signaling can upregulate the expression of certain genes among this group, forming positive feedback loops that reinforce cancer-related phenotypes.
The overexpression of these genes and activation of the PI3K-Akt pathway are interconnected in promoting gastric tumorigenesis and progression through their abilities to upregulate each other and mediate downstream oncogenic effects. Targeting this signaling nexus may have therapeutic potential in GC. The results of our investigation into these genes using DrugBank indicate that there are currently no drugs approved by the FDA targeting CTGF, THBS1, or WISP1. However, a few approved drugs do target FN1 and IL-6, such as Ocriplasmin (targeting FN1), and Foreskin fibroblast, Binimetinib, and Siltuximab (targeting IL-6). These drugs are suggested for further investigation as potential new therapeutic options for EC patients. Among these approved drugs, only Siltuximab has a single target (IL-6), indicating specific action and potentially fewer side effects. Therefore, Siltuximab could be considered a suitable choice for EC patients.
The exploration of pharmacotherapeutic agents targeting hub genes holds promise for personalized treatment strategies. While limited FDA-approved drugs targeting FN1 and IL6 were identified, the lack of approved agents for CTGF, THBS1, and WISP1 underscores an unmet therapeutic need. The multitarget nature of the drugs introduced in this study necessitates in-depth evaluation to assess potential off-target effects. Our findings advocate for future studies that delve into the mechanistic underpinnings of the identified hub genes and associated pathways. The integration of multi-omics data, coupled with experimental validation, could shed light on their roles in GC initiation, progression, and therapeutic response. Developing targeted therapies addressing these hub genes could potentially revolutionize treatment strategies for GC patients. Despite the comprehensive nature of our study, limitations such as the reliance on bioinformatics predictions and the lack of experimental validation merit consideration. Future investigations should encompass functional assays, animal models, and clinical validation to confirm the roles of these hub genes in gastric cancer pathogenesis and treatment response