Enhanced expression of HMMR in ccRCC is linked to the cancer progression and adverse clinical outcomes.
According to the TCGA database, we downloaded and integrated raw mRNA expression datasets of ccRCC, comprising a total of 614 samples, with 72 normal samples and 542 tumor samples. The findings indicated that there are notable differences in the expression levels of HMMR between normal and cancerous tissues, with a marked elevation observed in the latter (Fig. 1A1-2). The survival analysis, encompassing overall survival, disease-specific survival, and progression-free interval, revealed that the low expression group had a significantly longer survival duration compared to the high expression group (Fig. 1B1-3). HMMR expression also had satisfactory specificity and sensitivity in the ccRCC diagnosis wth AUC = 0.867 (Fig. 1B4). Combined with clinical data and HMMR expression levels, univariate and multivariate Cox regression models were established. Forest maps showed that HMMR was a risk factor for ccRCC patients in both univariate and multivariate analyses (Fig. 1C). Moreover, we found significant correlations between HMMR expression and patient characteristics such as gender, pathological grade, clinical stage, T stage, and M stage with HMMR expression increasing in tandem with advanced stages (Fig. 1D). This indicates a strong association between high HMMR expression and poor clinical outcomes. Additionally, Western blot analysis of 10 ccRCC samples from our center confirmed that cancerous tissues had significantly higher HMMR protein levels than non-cancerous tissues (Fig. 1E).
In summary, HMMR expression is up-regulated in the cancerous tissues of ccRCC patients, and this abnormal expression shows a positive correlation with the progression of ccRCC and predicts unfavorable clinical outcomes.
HMMR plays a significant role in the progression of ccRCC.
Through regression analysis of HMMR expression across all our samples, we discovered that different clinical indicators in ccRCC contribute to varying extents to the overall scoring process (Fig. 2A). Additionally, predictive analyses were conducted for 1-year and 3-year OS models (Fig. 2B), revealing a close correlation between the predicted OS and the observed OS, thereby validating the nomogram model's robust predictive performance. Expanding our investigation, we leveraged drug sensitivity data from the GDSC database to forecast chemotherapy sensitivity. This exploration further illuminated a significant relationship between HMMR and the sensitivity to several common anticancer drugs, including Nutlin-3a, AG-014699, AICAR, AZD-0530, AZD-2281, and A-443654 (Fig. 2C), suggesting HMMR may act as a potential factor influencing drug resistance.
Considering the tumor microenvironment (TME) has an important impact on aspects such as cancer diagnosis, patient survival, and responsiveness to clinical treatment, our study delved into the relationship between HMMR expression levels and the extent of immune cell infiltration within TCGA dataset. Our findings revealed a remarkable positive association between HMMR and certain immune cell populations integral to antitumor immunity, including cytotoxic CD8 + T cells and T follicular helper cells (Fig. 2D). Of note, a particularly striking finding was the positive correlation between heightened HMMR expression and an increased infiltration of T follicular helper cells, with a correlation coefficient (r) of 0.186 (Fig. 2E). This suggests that elevated HMMR levels coincide with an enrichment of helper T cells, which are vital for effective antibody responses and fostering an immunologically active TME. Such correlations provide some basis for further understanding the complex interaction between HMMR expression and the composition of the immune infiltrate, potentially informing future strategies for immunomodulatory therapies in cancer treatment.
To explore the mechanism of HMMR on tumor development, we also performed single-cell analysis of HMMR. Downloading data from GSE207493, we included 19 samples and utilized the t-SNE algorithm to cluster the cells, resulting in 21 distinct subtypes (Fig. 2F). Applying the R package SingleR for cell type annotation, we annotated these 21 clusters into nine cell categories, such as T cells, tissue stem cells, neurons, among others (Fig. 2G). The expression pattern of HMMR across these nine cell types was further depicted in Fig. 2H and 2I, shedding light on its cellular heterogeneity and potential roles in different cell populations.
HMMR regulates tumor growth in vivo and biological activities in vitro of ccRCC.
By conducting RT-qPCR and Western blot analyses on several RCC cell lines and HK-2, we found that both the transcriptional and protein levels of HMMR were notably higher in 786-O and 769-P cells than in normal cells (Fig. 3A). To reduce HMMR expression, we transfected these cells with small interfering RNAs targeting HMMR (siHMMR-1 and siHMMR-2) and confirmed a substantial decrease in HMMR expression at both mRNA and protein levels in 786-O and 769-P cells (Fig. 3B). Following HMMR knockdown, the migratory capability (Fig. 3C1), cellular viability (Fig. 3C2), and the efficiency of colony formation (Fig. 3C3) for these cells were notably reduced in vitro. To investigate the oncogenic potential of HMMR further, we established xenograft tumor models by subcutaneously injecting 786-O cells that stably expressed shHMMR or a negative control (shNC) into the axillary region of nude mice. The experimental results showed that HMMR knockdown significantly diminished the weight of 786-O xenograft tumors (Fig. 3D, E).
Moreover, we used an overexpression plasmid to overexpress HMMR in cells, confirming the efficiency of overexpression (Fig. 3F). Cells with HMMR overexpression exhibited significantly enhanced migratory ability (Fig. 3G1), proliferation (Fig. 3G2), and colony formation (Fig. 3G3) in vitro.
Collectively, these findings suggest that HMMR positively regulates the migration and proliferation of ccRCC, highlighting its crucial role in ccRCC progression.
miR-9-5p is postulated to regulate HMMR expression and influence ccRCC cells proliferation and migration.
Available literature underscores the key regulatory functions of miRNAs in the onset and advancement of diverse malignancies including RCC [23]. Consequently, we hypothesized that the oncogenic function of HMMR in ccRCC might be modulated by upstream miRNAs. To test this, we retrieved the miRNA expression profiles for ccRCC from TCGA database and performed differential expression analysis using three R packages: DEseq2, limma, and edgeR, to identify miRNAs that are under-expressed in tumor tissues. Integrating these findings with the predictive interactions from the ENCORI online tool, we constructed a Venn diagram, which led us to select 2 candidate miRNAs, miR-509-3p and miR-9-5p (Fig. 4A). Notably, miR-9-5p was expressed at a lower level than HK-2 in the RCC cell lines we used, with a particularly pronounced downregulation observed in 786-O and 769-P cells (Fig. 4B).
Subsequently, we introduced miR-9-5p mimics or negative controls (NCs) into 786-O and 769-P cells. Through RT-qPCR and Western Blotting, we verified that the transfection of miR-9-5p mimics markedly decreased HMMR expression compared to the NC group (Fig. C).
For further study of the carcinogenic potential of miR-9-5p in ccRCC cells, we carried out additional cellular assays after the transfection with negative controls or miR-9-5p mimics. Transwell migration assays demonstrated that there was a notable decrease in the migration capability of both 786-O and 769-P cells (Fig. 4D1). CCK-8 assays showed that cell proliferation was significantly inhibited in cells miR-9-5p mimics group compared to control group (Fig. 4D2). Colony formation assays also showed that the cloning efficiency of cells decreased significantly after overexpression of miR-9-5p (Fig. 4D3). In conclusion, the above functional assays imply that miR-9-5p may exert an antitumor effect in ccRCC by suppressing ccRCC cells proliferation and migration.
miR-9-5p exhibits targeted interaction with HMMR to mediate biological activity of ccRCC.
We had uncovered the oncogenic potential of HMMR and the significant tumor-suppressive effect exerted by miR-9-5p. To explore the intrinsic mechanism, a series of rescue experiments involving HMMR and miR-9-5p were conducted. HMMR overexpression could partially restore the decreased HMMR protein expression induced by miR-9-5p mimics, as confirmed by Western blot analysis (Fig. 5A). Functional assays following this setup, including colony formation assays (Fig. 5B) and CCK-8 assays (Fig. 5C), suggested that elevated HMMR expression could partially reverse the reduction in proliferation caused by miR-9-5p. Similarly, Transwell migration assays (Fig. 5D) indicated that HMMR overexpression could also rescue the migratory capacity decrease induced by miR-9-5p.
To conclusively validate whether HMMR is a direct target of miR-9-5p, the WT and Mut 3’-UTRs of HMMR mRNA were separately cloned into the pmirGLO vector (depicted in Fig. 5E). The outcome revealed a significant decrease in the relative luciferase activity of the WT group after miR-9-5p was overexpressed in 293T cells (Fig. 5F), confirming the direct interaction and regulation of HMMR by miR-9-5p.
HMMR facilitates the progression of ccRCC via the activation of both EMT and JAK/STAT signaling pathway.
EMT is a critical process involved in the acquisition of invasive properties by cancer cells, affecting their motility and migratory abilities [28]. By knocking down HMMR expression, at the protein level, we observed a decrease in N-cadherin, Slug, and Vimentin alongside an increase in E-cadherin, suggesting a potential involvement of HMMR in the EMT process of ccRCC cells. Matrix metalloproteinases (MMPs), closely tied to the EMT process and responsible for degrading extracellular matrix components to facilitate cancer cell invasion and migration, were also found to be downregulated, specifically MMP9 (Fig. 6A). These results indicate that HMMR might promote the progression of ccRCC by enhancing EMT process.
Furthermore, we employed GSEA to further elucidate the specific signaling pathways implicated by HMMR, thereby delving into the underlying molecular mechanisms by which HMMR influences tumor initiation and progression. Our findings indicated a notable enrichment of the JAK/STAT signaling pathway in association with HMMR (Fig. 6B). Recent study has shown that JAK1/STAT1 is involved in the progression and immunotherapy evasion of ccRCC, thus the pathway was tested in this research [29]. By transfecting 786-O and 769-P cells with either HMMR-overexpressing plasmids or empty vectors, and subsequently assessing the expression levels of JAK1, STAT1, and phosphorylated STAT1 (p-STAT1) proteins, we found that the overexpression of HMMR led to increased expression of both JAK1 and p-STAT1 proteins in these cells (Fig. 6C). This indicates that HMMR upregulates key components of the JAK/STAT signaling pathway. To further validate whether HMMR promotes the progression of ccRCC via the JAK/STAT signaling pathway, we cultured these cells in media containing Ruxolitinib, a selective inhibitor of JAK. The outcomes demonstrated that Ruxolitinib effectively suppressed the migratory (Fig. 6D) and proliferative capacities (Fig. 6E) of cells that had been induced by HMMR overexpression.
In summary, these findings suggested that HMMR facilitated the progression of ccRCC through the activation of both EMT and JAK1/STAT1 signaling pathway.