MLST8 is a potential biomarker for ccRCC according to TCGA
We first exploited the TCGA database to analyze MLST8 mRNA expression and its association with ccRCC prognosis. As shown in Fig. 1a, MLST8 was significantly upregulated in ccRCC samples compared to normal tissues at the transcriptional level (p < 0.0001). We next evaluated this association with clinical-pathological characteristics. Although there was no difference between MLST8 expression and lymphatic metastasis, MLST8 was correlated to tumor stage M stage (Fig. 1b-e). Because the N1 stage samples from TCGA were small, it was insufficient to demonstrate a correlation between MLST8 and N metastasis. However, a strong correlation was identified between the MLST8 expression and the overall survival of ccRCC patients (Fig. 1f, p = 0.0248). Therefore, we assumed that the non-significance between MLST8 and distant metastasis was potentially due to the limited sample size, necessitating future validation.
MLST8 is upregulated in ccRCC specimens and predicts poor clinical outcomes in ccRCC patients
To confirm the correlation between MLST8 expression and ccRCC progression, we performed immunohistochemistry (IHC) to assess the MLST8 protein expression in 79 human ccRCC specimens. Substantially overexpressed MLST8 protein was found in ccRCC specimens compared to adjacent normal tissue (Supplementary Table S2, p < 0.0001), and results showed a significant increase in the expression levels of MLST8 as ccRCC progressed to an advanced stage (Fig. 2a, b). The association between MLST8 expression and clinical pathological characteristics are shown in Supplementary Table S2. The over-expression of MLST8 was strikingly correlated to the T stage (p = 0.009), TMN stage (p = 0.058), and Furman grade (p <0.0001). However, there were no association between MLST8 expression levels and a patient’s age, gender, and size. We developed Kaplan-Meier curves on 75 patients with a log-rank test for OS to elucidate the relationship between MLST8 expression and patients’ survival in ccRCC. Our results reveal that a high expression of MLST8 is associated with poor OS (Fig. 2c, p =0.0183). These results suggest that MLST8 is a potential prognostic marker in ccRCC.
FBXW7 interacts with MLST8
A previous study was performed quantitative mass spectrometry using FBXW7 knockout (KO) HCT116 cells [23]. Enrichment of MLST8 protein in the nuclear fraction of the HCT116 FBXW7-KO cells could be a binding partner for FBXW7, and most likely, the alpha isoform that resides in the nucleus (Figure 3a). To verify that FBXW7 is a bona fide MLST8 interactor, we first examined whether FBXW7 can interact with MLST8 cells. FLAG-FBXW7 and Myc-MLST8 constructs were co-expressed in 293T cells. Cells were subsequently harvested for co-immunoprecipitation (Co-IP) with the anti-FLAG antibody. As shown in Figure (Figure 3b), Myc-MLST8 was immunoprecipitated by FLAG-FBXW7, suggesting an exogenous interaction between these two proteins. Besides, a reciprocal Co-IP assay was performed using lysates of 293T cells that were co-transfected with FLAG-MLST8 and Myc-FBXW7 constructs. The results indicated that FLAG-MLST8 was also able to immuneprecipitate Myc-FBXW7 (Figure 3c). Then experiments further show that FLAG-FBXW7 was capable of immunoprecipitated endogenous MLST8 in 786-O cells (Figure 3d). We explored whether endogenous MLST8 and FBXW7 have an immediate interaction with each other. Immunoprecipitation using the anti-MLST8 antibody was performed using cell lysates prepared from 786-O cells. As shown in Figure 3e, endogenous FBXW7 was immunoprecipitated by MLST8, suggesting an endogenous interaction between these two proteins. Similarly, endogenous MLST8 was immunoprecipitated by FBXW7 (Figure 3f). To investigate whether FBXW7 co-localizes with MLST8 in vivo, 786-O, and A498 cells were transfected with FLAG-MLST8 and Myc-FBXW7, immunostained and visualized by confocal microscopy. As shown in Figure 3g, FBXW7 and MLST8 were co-localized in the nucleus. Taken together, these results indicate that FBXW7 forms a complex with MLST8 in cells.
MLST8 is degraded and ubiquitinated by tumor suppressor FBXW7
We next sought to investigate the mechanism by which FBXW7 regulates the protein levels of MLST8. Myc-FBXW7α and Flag-MLST8 were co-transfected in 293T cells, followed by treatment with DMSO and MG132, respectively. Although Myc-FBXW7α promoted the degradation of MLST8, this effect was inhibited by the treatment with the proteasome inhibitor MG132 (Figure 4a). We used CRISPR-Cas9 to knockout endogenous copies of FBXW7 in 786-O and A498 cells with two FBXW7-specific sgRNAs, resulting in an increase in MLST8 protein levels (Figure 4b) but no effect on the mRNA expression (Figure 4c). Further results showed that the depletion of FBXW7 significantly extends the half-life of endogenous MLST8 protein and stabilizes the levels of the MLST8 protein (Figure 4d-4e). Furthermore, when 786-O cells were transfected with three isoforms (α, β, and γ) of FBXW7, all resulted in a marked reduction in the protein levels of MLST8 in a dose-dependent manner (Figure 4f). Since FBXW7 is an ubiquitinating enzyme, we wondered whether FBXW7 can ubiquitinate and downregulate MLST8 protein levels. As shown in Figure 4g, ectopic expression of FBXW7-WT, but not the FBXW7-△F-box mutant (catalytic inactive), decreased the protein levels of co-expressed MLST8 in a dose-dependent manner, suggesting that it’s the ubiquitination activity of FBXW7 to promote MLST8 destabilization. Conversely, the depletion of FBXW7 using CRISPR-Cas9 knock out decreased MLST8 ubiquitination levels (Figure 4h), indicating that FBXW7 is responsible for MLST8 ubiquitination. Additionally, MLST8 protein was robustly polyubiquitinated in a dose-dependent manner when FBXW7-WT was co-expressed. By contrast, minimal or no MLST8 polyubiquitination was observed in FBXW7-△F-box expressing cells (Figure 4i). In sum, these data demonstrate that the FBXW7 E3 ubiquitin ligase complex regulates MLST8 protein stability through the ubiquitin-dependent proteasomal degradation pathway in ccRCC cell lines.
Thr50-dependent is required for FBXW7-mediated degradation of MLST8
The FBXW7-binding consensus motif (T/S)PXX(S/T/D/E) has been identified in several FBXW7 substrates, including cyclin E, c-Myc, c- Jun [9, 10], and OASIS [23]. When we examined the amino acid sequence of MLST8, we discovered that MLST8 harbors one perfectly matched (50-TPDRS-54) FBXW7-binding motif (Figure 5a-b). FBXW7 often recognizes phosphorylated Ser/Thr residues in the CPD motif. Therefore, we speculated that the regulation of MLST8 by FBXW7 might be mediated by the phosphorylation of Thr50 in its CPD. To examine whether this region is required for the FBXW7–MLST8 interaction, we generated MLST8 mutants in which the acidic residues T50 and S54 residues were mutated to Alanine. 293T cells were co-transfected with FBXW7α and MLST8-WT or MLST8 mutant. Although FBXW7 coimmunoprecipitated MLST8-WT at a similar level, both MLST8 mutant completely lost its FBXW7-binding capability (Figure 5c). These data indicate that the 50-TPDRS-54 motif is necessary for the FBXW7–MLST8 interaction. Next, we sought to identify whether the 50-TPDRS-54 motif is required for the FBXW7-mediated degradation of MLST8. As shown in Figure 5d, while FBXW7 efficiently targets MLST8-WT for degradation, the MLST8-T50A/S54A mutant displayed resistance to FBXW7-mediated degradation, indicating that the 50-TPDRS-54 motif is crucial to FBXW7-mediated MLST8 degradation. Furthermore, the T50A/S54A mutation prolonged the half-life of MLST8 protein (Figure 5e-f). Next, in vivo ubiquitination assays suggest that FBXW7 robustly enhanced the polyubiquitination of MLST8-WT, but not the MLST8-T50A/S54A mutant (Figure 5g). Consistent with the protein degradation, the T50A/S54A mutation largely diminished FBXW7 induced MLST8 ubiquitination. Therefore, these data demonstrate that the 50-TPDRS-54 motif functions as an MLST8 degron and is essential for FBXW7 binding and degradation through the ubiquitin-proteasome pathway.
Activated CDK1 induces degradation of MLST8 and decreases the binding with FBXW7 and MLST8
FBXW7-mediated degradation requires the phosphorylation of the first Ser/Thr in the CPD motif of FBXW7 target proteins by a variety of kinases, including GSK3β [24], CDK1/2 [22], CDK4, or CDK5 [25]. To identify the kinase involved in the phosphorylation of T50 in MLST8, we first analyzed the surrounding amino acid sequence and found that Thr50 in MLST8 perfectly matches the general CDK1/2 consensus motif (S/TPXK/R) (26). WT-MLST8 expressed in 293T cells treated with phosphatase and proteasome inhibitors was detected by an anti-p-T50-MLST8 antibody, but this was not detected when using the T50A-MLST8 mutant (Figure 6a). Thus, the Thr50 of exogenous MLST8 is phosphorylated in vivo. Furthermore, we detected endogenous MLST8 and its phosphorylation on Thr50 in 786-O cells transfected with MLST8 Control (Figure 6b), suggesting that endogenous MLST8 was phosphorylated at Thr50 in 786-O cells. The CPD motif in MLST8 (Thr50-Pro51-Asp52-Lys53-Ser54) corresponds to the consensus motif for CDKs, which is Ser/Thr-Pro-X-Lys/Arg (Figure 6C). To identify the kinase responsible for phosphorylation of MLST8 at residue Thr50, we performed an in vitro phosphorylation assay using purified GST-MLST8 protein and recombinant cyclin-CDK complexes. Phosphorylation of Thr50 in mLST8 by a variety of kinases, including CDK5, CDK4, and CDK2, was not detected, but phosphorylation on a control RB protein was at the same efficiency (Figure 6d-e). CDK1 efficiently phosphorylated MLST8 at Thr50, while CDK2 slightly resulted in phosphorylation (Figure 6d). We also examined whether recombinant GSK3β phosphorylates GST-MLST8 in vitro (Figure 6f). We determined that GSK3β did not phosphorylate Thr50 in MLST8 in vitro and is not the kinase responsible for the CPD motif of MLST8. Furthermore, we investigated whether phosphorylation of Thr50 in MLST8 was required for its recognition by FBXW7 using a GST pull-down assay. Purified GST-WT-MLST8 or GST-T50A-MLST8 was phosphorylated by CDK1 and incubated with cell lysates expressing FLAG-FBXW7. MLST8 in the presence of CDK1 migrated more slowly than the unphosphorylated form of MLST8, even when Thr50 was mutated to alanine (T50A). we also found that FBXW7 bound to GST-WT-MLST8 was detected by an anti-p-T50-MLST8 antibody after treatment with CDK1, but did not bind to GST-MLST8 without CDK1(Figure 6g). Therefore, these results suggest that CDK1 phosphorylation of residue Thr50 in MLST8 is responsible for its recognition by FBXW7.
MLST8 is an important mediator of FBXW7 inactivation-induced cell proliferation, migration and invasion in vitro
Accumulating shreds of evidence showed that MLST8 expression was upregulated in various types of human cancers. This evidence drove us to explore whether MLST8 is involved in FBXW7 depletion-induced increase in cell proliferation, migration, and invasion. As determined by the CCK-8 assay, the growth rate of MLST8-depleted 786-O and A498 cells was slower than the control cells. In contrast, the depletion of FBXW7 could significantly enhance cell proliferation, and the phenotypes could be rescued by MLST8 co-depletion (Figure 7a-b). We observed similar effects as the cell proliferation assay, in which the increase of migration and invasion in 786-O and A498 cells by FBXW7 depletion was partly diminished by MLST8 inactivation (Figure 7C-D). Thus, these results suggest that MLST8 is an essential mediator of FBXW7 inactivation-induced cell proliferation, migration, and invasion.
MLST8 and FBXW7 Protein levels negatively correlate in human renal cancer specimens
Given the critical role of FBXW7 and MLST8 in ccRCC, we analyzed the expression of FBXW7 and MLST8 by IHC on a tissue microarray (TMA) in 79 pairs of renal cancer samples with clinical follow-up information and observed that both are mainly expressed in the nucleus which is consistent with the result of immunofluorescence. Statistical analysis revealed that MLST8 was remarkably overexpressed in renal cancer tissues compared to normal tissue. In contrast, FBXW7 was expressed at a lower level in tumor tissues (Figure 8a). The clinical and pathologic characteristics of the patients are shown in Supplementary Table. S2. In the 79 paired ccRCC cases, MLST8 protein was greatly overexpressed in 60.0% of tumor tissues, and FBXW7 was found to be under-expressed in 68.4% of tumor tissues (Figure 8b). Moreover, statistical analysis indicated that MLST8 was negatively correlated with FBXW7 expression in this cohort of patient samples (r=-0.3124, p=0.0051) (Figure 8c). This result suggested that FBXW7-mediated MLST8 decrease may also restrain renal cancer progression. Further, the univariate and multivariate analyses were used to determine the independent prognostic factors for ccRCC patients (Supplementary Table S3). In univariate analysis, the MLST8 expression level instead of FBXW7, was found to be significantly associated with the OS (p =0.006). TNM stage (p < 0.0001) and Fuhrman grade (p = 0.001) were also correlated significantly with OS, nevertheless other factors such as tumor size (p = 0.058) and age (p = 0.871) were also uncorrelated significantly with OS. Moreover, multivariate analysis showed that the TNM stage (p = 0.000), tumor size (p = 0.050) were proven to be independent predictors of OS for ccRCC patients. Simultaneously, other variables, including age and Fuhrman grade, did not contribute to overall survival independently (P > 0.05).