Front Oncol. 2025 ;15 1577114
Background: Phospholipase C η1 (PLCH1), a member of the phospholipase C superfamily, has been implicated in the development of multiple cancers. However, its specific role in breast cancer progression, its association with clinicopathological features, and its prognostic significance remain unclear.
Methods: PLCH1 expression was analyzed across multiple tumor types using the TNMplot database, which integrates RNA-seq, microarray, and normalized data from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Gene Expression Omnibus (GEO), encompassing 40,442 tumor and 15,648 normal samples. Differential expression analysis was performed using boxplots and statistical tests to assess significance. DNA methylation and survival analyses were conducted using TCGA data, with Kaplan-Meier curves and Cox regression to evaluate prognostic value. Functional enrichment analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, were performed on differentially expressed genes using the clusterProfiler package. Mutation analyses were conducted using mutation annotation format (MAF) files, and pathway activities were correlated with PLCH1 expression via single-sample GSEA (ssGSEA). Experimental validation included immunohistochemistry (IHC) on 100 breast invasive ductal carcinoma samples, real-time quantitative PCR (RT-qPCR), and Western blotting. PLCH1 knockdown functional studies assessed cell proliferation and signaling pathways.
Results: PLCH1 was significantly overexpressed in various cancers, including breast cancer, compared to normal tissues. PLCH1 expression was strongly correlated with the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) in breast cancer tissues, further linking PLCH1 to poor prognosis and adverse patient outcomes. Functional studies revealed that PLCH1 was highly expressed in breast cancer cell lines, and PLCH1 knockdown significantly inhibited cell proliferation, induced cell cycle arrest, and reduced cyclin-dependent kinase 1 (CDK1) expression in BT-474 cells. Mechanistically, PLCH1 silencing downregulated early growth response 1 (EGR1) expression by suppressing the extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling pathway, impairing tumor cell proliferation.
Conclusions: PLCH1 was overexpressed in breast cancer and was associated with worse patient outcomes. Its role in promoting cell proliferation via the ERK1/2-EGR1 axis highlighted PLCH1 as a potential therapeutic target for breast cancer. These findings offer new insights into the molecular mechanisms underlying breast cancer progression and suggest promising avenues for targeted therapy development.
Keywords: ERK1/2; PLCH1; apoptosis; breast cancer; cell cycle