Abstract

Objective Casein kinase 2 interacting protein-1 (CKIP-1) has exhibited multiple functions in regulating cell proliferation, apoptosis, differentiation, and cytoskeleton. CKIP-1 also plays an important role as a critical regulator in tumorigenesis. The aim of this study is to further examine the function of CKIP-1 in glioma cells. Methods The expression level of CKIP-1 protein was determined in gliomas tissues and cell lines by immunohistochemistry stain and western blotting while the association of CKIP-1 expression with prognosis was analyzed by Kaplan-Meier method and compared by log-rank test. CKIP-1 was overexpressed or silenced in gliomas cell lines. CCK-8, colony formation assay, and BrdU incorporation assay were used to determine cell proliferation and DNA synthesis. Cell cycle and apoptosis rate were determined with fluorescence-activated cell sorting (FACS) method. Then, expression of key members in AKT/GSK3β/β-catenin pathway was detected by western blot analysis. Results In the present study, we reported new evidence that CKIP-1 was reversely associated with the proliferation of glioma cells and survival in glioma patients. Additionally, the overexpressed CKIP-1 significantly inhibited glioma cell proliferation. Further experiments revealed that CKIP-1 functioned through its antiproliferative and proapoptotic activity in glioma cells. Importantly, mechanistic investigations suggested that CKIP-1 sharply suppressed the activity of AKT by inhibiting the phosphorylation, markedly downregulated the phosphorylated GSK3β at Ser9, and promoted β-catenin degradation. Conclusions Overall, our results provided new insights into the clinical significance and molecular mechanism of CKIP-1 in glioma, which indicated CKIP1 might function as a therapeutic target for clinical treatment of glioma.

Highlights

  • In the central nervous system, gliomas are the most common type of aggressive primary solid tumor, accounting for ∼80% of primary malignant brain tumors

  • We found Casein kinase interacting protein-1 (CKIP-1) staining clearly was localized to the cytoplasm and nuclei in nontumor brain tissue (Figure 1(a)) and generally higher than glioma tissue

  • CKIP-1 depletion (U87-RNAi) resulted in an accumulation in S phase in greater numbers but reduced that in G0/G1 phase than U87-Ctrl cells (all p < 0.01; Figure 3(b), right). These results demonstrated that CKIP-1 arrested glioma cells at the G0/G1 phase which control that cells do not exit from G1 phase and enter into S phase of the cell cycle, while depletion of CKIP-1 reversed the accumulation in the G0 /G1 phase and allowed the glioma cells to enter into S phase, suggesting that CKIP-1 may function through its antiproliferative activity in glioma cells

Read more

Summary

Introduction

In the central nervous system, gliomas are the most common type of aggressive primary solid tumor, accounting for ∼80% of primary malignant brain tumors. Malignant glioma is characterized by invasion into surrounding brain tissues, rapid proliferation, and aberrant angiogenesis [2]. These biological characteristics lead to unclear boundaries between tumors and surrounding brain tissues and the glioma cells usually spread deep into the brain which pose a great challenge to surgeons. Multiple therapies include maximum safe resection, followed by chemotherapy, radiation, and immunotherapy, the outcome of patients diagnosed with high-grade glioma remains unsatisfied, with median survival of less than 15 months [3, 4]. It is vital to investigate the specific molecular mechanism that regulate the progression of glioma and identify the novel molecular therapeutic targets to aid in development of glioma therapies

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call