GGCT is overexpressed in PTC tissues and cell lines.
To explore the expression pattern of GGCT in PTC, we first detected its expression in 27 pairs of PTC tissues and paracancerous tissues by RT-qPCR. As illustrated in Figure 1A, PTC tissues exhibited prominently elevated GGCT mRNA expression as compared to the corresponding paracancerous tissues. These data were also confirmed in the TCGA database, in where GGCT was significantly higher in tumor tissues than in normal ones for both unpaired (Supplementary Figure 1A) and paired (Supplementary Figure 1B) simple comparisons. Additionally, we further assessed GGCT protein level in 6 pairs of PTC and non-cancerous tissues using western blot analysis which showed most PTC tissues possessed greater GGCT expression than the normal controls (Figure 1B and 1C). Similar findings were also obtained with paraffin-embedded sections including 178 PTC and 82 normal thyroid tissues by IHC. As shown in Figure 1D and 1E, higher expression of GGCT was observed in the PTC tissues compared with the normal counterparts. (P<0.05). Besides, we measured a range of thyroid-derived cell lines for GGCT expression, and as expected, high levels of GGCT expression were visible in all three PTC cell lines but not in the normal thyroid follicular epithelial cell line (Nthy-ori-3-1) (Figure 1F and 1G). As GGCT has been estimated to have an elimination half-life in serum of up to 30 hours (https://web.expasy.org/cgi-bin/protparam/), we compared the blood content of GGCT preoperative and 24 hours postoperative from the same patient (n=26), and the results clearly indicated the serum GGCT level was noticeably reduced after surgical intervention (Figure 1H). Together these results strongly suggest that GGCT is upregulated in the PTC tissues and cell lines.
High-level of GGCT correlates with unfavorable clinicopathological characteristics and worse outcomes.
We focused on whether GGCT overexpression was responsible for worse PTC prognosis. Based on the IHC staining score, the patients were classified into the low-GGCT (n=67) and high-GGCT expression group (n=111). The Fishers exact or chi-square test showed that GGCT expression was strongly associated with tumor histological type (p<0.001), extrathyroidal extension (p=0.035), primary tumor classification (p=0.004), and TNM stage (p=0.002) (Table 1). In addition, Kaplan-Meier survival curve revealed a shorter disease-free survival (DFS) time in high-GGCT expression group than in low-GGCT expression group (Supplementary Figure 2). These results infer that GGCT is closely correlated with tumor malignant properties and decreases DFS in PTC patients.
Table 1
Relationship between GGCT expression and clinicopathological characteristics in 178 PTC tissues.
Clinicopathologic parameters | n | GGCT expression | p |
| | low | high | |
All cases | 178 | 67 | 111 | |
Age | | | | 0.848 |
≤ 55 | 60 | 22 | 38 | |
>55 | 118 | 45 | 73 | |
Gender | | | | 0.159 |
male | 69 | 20 | 49 | |
female | 119 | 47 | 72 | |
Histological type | | | | <0.001 |
classical | 106 | 39 | 67 | |
follicular | 28 | 19 | 9 | |
tall cell | 44 | 9 | 35 | |
Multifocality | | | | 0.218 |
unifocal | 129 | 45 | 84 | |
multifocal | 49 | 22 | 27 | |
Extrathyroidal extension | | | | 0.035 |
no | 133 | 56 | 77 | |
yes | 45 | 11 | 34 | |
T classification | | | | 0.004 |
T1-T2 | 117 | 53 | 64 | |
T3-T4 | 61 | 14 | 47 | |
Lymph node metastasis | | | | 0.395 |
no | 87 | 30 | 57 | |
yes | 91 | 37 | 54 | |
TNM stage | | | | 0.002 |
I+II | 128 | 57 | 71 | |
III+IV | 50 | 10 | 40 | |
Braf-V600E mutation | | | | 0.975 |
no | 72 | 27 | 45 | |
yes | 106 | 40 | 66 | |
Knockdown of GGCT alleviates the proliferation, migration and invasion of PTC cells in vitro and in vivo.
Given the findings that upregulation of GGCT was associated with poorly clinicopathological characteristics and worse outcomes in PTC patients, we postulated that GGCT might act as a tumor facilitator in PTC. To assess the effect of GGCT on the biological behaviors of PTC cells, we performed the RNA interference experiment with three distinct shRNA duplexes. Among them, shRNA-3 exhibited the best knockdown efficiency, as indicated by WB analysis, and thus selected for the subsequent investigations (Figure 2A). Firstly, we assessed the impact of GGCT on cell growth through the CCK-8 assay. As shown in Figure 2B and 2C, GGCT knockdown remarkably inhibits the proliferation of K1 and BCPAP in a time-dependent manner. We next evaluated the effect of GGCT on cell migration and invasion via wound healing and Matrigel Transwell assays. The wound healing assay demonstrated that silencing of GGCT significantly retarded the closure of the wound gap after 24 hours and 48 hours (Figure 2D and 2E). Also, shRNA-mediated abrogation of GGCT impeded the matrix penetration capability of K1 and BCPAP cell lines (Figure 2F and 2G).
Since epithelial-mesenchymal transition (EMT) has previously been proved to be critical for the survival and invasiveness acquisition of cancer epithelial[26]. We questioned whether interference with GGCT expression inhibits cell proliferation, migration and invasion by reversing the EMT phenotype. Indeed, after stably downregulation of GGCT in PTC cells, K1 and BCPAP cells underwent several morphologic alterations from mesenchymal spindle-like phenotype to epithelial polarized phenotype in contrast to the control cells (data not shown). Subsequently, the EMT-related proteins were examined by WB analysis, and the results showed a downregulation of mesenchymal markers (N-cadherin, CD44, MMP-2, MMP-9) and a concomitant upregulation of epithelial marker (E-cadherin) in sh-GGCT cells, heralding the EMT-promoting role of GGCT in PTC (Figure 2H).
To determine whether GGCT is required for in vivo tumor growth and metastasis, the nude mouse subcutaneous xenotransplant tumor model and the tail vein–lung metastasis model was performed. As pointed out in Figure 3A-C, GGCT-knockdown K1 cells exhibited an evidently decreased tumor volume and tumor weight 3 weeks post-modeling compared with the control group. Similar results were yielded by the xenografts constructed using BCPAP cell lines which were stably infected with sh-GGCT or sh-NC lentivirus (Figure 3D-F). Moreover, the efficiencies of GGCT knockdown in vivo and the EMT-associated markers were assessed by WB analyses, which proved the silencing of GGCT contributed to the reduction of tumor growth and reversion of EMT process (Supplementary Figure 3). As for the experimental pulmonary metastasis assay, sh-GGCT-K1 or sh-NC-K1 cells expressing firefly luciferase were administered via tail vein injection. The successful aggregation of tumor cells into the lung in both sh-GGCT and sh-NC groups was confirmed at day 0 by bioluminescence imaging (Figure 3G). With extended observation time, the bioluminescence signals in the sh-NC-K1 group rapidly increased and all mice in this group were forced to be sacrificed due to the tumor-associated cachexia up to 3 weeks post injection (Figure 3G-H). Surprisingly, none of the positive signals were captured in the sh-GGCT-K1 group throughout the observation period, demonstrating an entire abrogation of the formation of lung metastatic foci by GGCT repression (Figure 3G-H). These were also corroborated by an ex vivo imaging of the visceral organs in combination with HE staining of lung tissues (Figure 3I-J). All above shed light on that abolishment of GGCT weakened the malignant potential of PTC cells in vitro and in vivo by reversing the EMT process.
MiR-205-5p can directly bound to the 3’UTR of GGCT and regulate GGCT expression.
Evidences support the crucial role of miRNAs in various cancers by inhibiting their target genes[27]. Interestingly, mir-205-5p, a potential tumor suppressor in multiple malignancies, was predicted to contain the binding site of the 3UTR region of GGCT, as obtained by bioinformatics (Figure 4A). Further exploration showed that decreased expression of GGCT was observed in K1 cells transfected with mir-205-5p mimics, while increased expression in mir-205-5p inhibitor groups (Figure 4B). To probe whether GGCT was the direct target of miR-205-5p, GGCT-3UTR with a wild type (WT) or mutated (MUT) miR-205-5p binding motif was constructed and subcloned into the dual-luciferase vector (Figure 4C). It was found that miR-205-5p reduced the luciferase activity of WT 3UTR-reporter, but not the MUT 3’UTR-reporter (Figure 4D). Moreover, biotin RNA-RNA pull-down assay revealed that GGCT mRNA were significantly enriched by using Bio-miR-205-5p-WT than Bio-miR-205-5p-MUT and Bio-NC (Figure 4E-F), indicating the interactions between miR-205-5p and GGCT.
MiR-205-5p reverses the pro-malignant phenotypes induced by GGCT dysregulation.
To corroborate that mir-205-5p reverses the GGCT-induced promotion of cell proliferation and invasion, pre-miR-205 was introduced into K1-GGCT cells by lentiviral overexpression. Overexpressing of miR-205 resulted in an attenuated proliferation ability of K1-GGCT cells (Figure 5A). Also, upregulation of miR-205 in K1-GGCT cells leads to the inhibition of migratory (Figure 5B-C) and invasive (Figure 5D-E) capacities, accompanying with increased expression of the epithelial markers and decreased mesenchymal markers (Figure 5F). To further confirm if miR-205-5p eventually restores GGCT-mediated tumorigenicity and metastasis in vivo, K1-GGCT cells with or without miR-205 overexpression were administrated into immune-deficient nude mice. Introduction of miR-205 not only repressed tumor growth (Figure 5G-H), but diminished the metastatic lesions in the metastatic model (Figure 5I-J). Altogether, these findings revealed that miR-205-5p suppressed the tumorigenicity and metastasis of PTC by targeting GGCT.
GGCT interacts with CD44 and inhibits the degradation of CD44.
Considerable evidence indicates that the adhesion molecule CD44 plays a vital role in mediating chemoresistance, cell proliferation and EMT[25]. As data preceding indicates that CD44 expression was positively influenced by GGCT, we were therefore intrigued by which mechanism this phenomenon happens. As depicted in Figure 6A, overexpression of GGCT significantly upregulated the CD44 protein, but leave no effect on the mRNA level of CD44. Subsequently, co-immunoprecipitation assays were conducted in K1 cell extracts. We co-expressed CD44-his with vector or GGCT-flag in HEK293T cells. We saw a co-precipitated CD44-his when GGCT-flag was pulled down (Figure 6B). Also, the interaction was further substantiated by reciprocal co-immunoprecipitations (Figure 6C). Consistently, by immunoprecipitating GGCT with anti-GGCT, CD44 protein was successfully precipitated, confirming the existence of an endogenous GGCT-CD44 complex in K1 cells (Figure 6D). Subsequently, a cycloheximide (CHX) chase experiment was conducted to explore the function of GGCT on CD44 stability by blocking de novo protein biosynthesis. Exposure to CHX results in obviously degradation of CD44 protein in HEK293T cells transfected with GGCT-vector, but not in cells transfected with GGCT (Figure 6E). Furthermore, to evaluate the relationship of GGCT and CD44 expression, we carried out an immunohistochemical analysis of human PTC simples, and observed that CD44 is positively correlated with GGCT protein levels (Figure 6F-G). Finally, an analysis of public databases demonstrated that high expression of CD44 is correlated with worse DFS of patients with PTC (Figure 6H). Collectively, these data signified that GGCT positively regulates CD44 expression by inhibiting its protein degradation.