Abstract

BackgroundSelective cyclooxygenase (COX)-2 inhibitors elicit anti-proliferative responses in various tumours, however the underlying anti-tumour mechanisms are unclear. Mutational inactivation of the tumour suppressor p53 gene is frequent in malignant gliomas. The role of p53 mutation in the anti-tumour responses of the selective COX-2 inhibitor celecoxib in human glioblastoma cells is unknown. In this study, we used human glioblastoma cells with various p53 status; U87MG (with high and low p53 functional levels), LN229 (functional p53) and U373MG (mutant p53) cells. Inhibition of p53 was achieved in U87MG cells transfected with E6 oncoprotein (U87MG-E6) and treated with pifithrin-α, a reversible inhibitor of p53 (U87MG-PFT). We investigated whether the anti-glioblastoma responses of celecoxib were p53-dependent, and whether celecoxib induced DNA damage leading to p53-dependent G1 cell cycle arrest, followed by autophagy or apoptosis.ResultsOur findings demonstrated that celecoxib concentration-dependently reduced glioblastoma cell viability, following 24 and 72 hours of treatment. Inhibition of functional p53 in glioblastoma cells significantly reduced the anti-proliferative effect of celecoxib. In U87MG cells, celecoxib (8 and 30 μM) significantly induced DNA damage and inhibited DNA synthesis, corresponding with p53 activation. Celecoxib induced G1-phase cell cycle arrest, accompanied with p21 activation in U87MG cells. Cell cycle progression of U87MG-E6 and U87MG-PFT cells was not affected by celecoxib. In parallel, celecoxib induced G1 cell cycle arrest in LN229 cells, but not in U373MG cells. Autophagy was induced by celecoxib in U87MG and LN229 cells, as shown by the significantly greater population of acridine orange-stained cells and increased levels of LC3-II protein (in comparison with non-treated controls). Celecoxib did not induce significant autophagy in U87MG-PFT, U87MG-E6 and U373MG cells, which lack functional p53. Regardless of p53 status, celecoxib caused no significant difference in apoptosis level of U87MG, U87MG-PFT, U87MG-E6 and U373MG cells.ConclusionOur findings reveal that p53 increases human glioblastoma sensitivity to celecoxib. Celecoxib inhibits glioblastoma cell viability by induction of DNA damage, leading to p53-dependent G1 cell cycle arrest and p53-dependent autophagy, but not apoptosis.

Highlights

  • Despite conventional therapy of surgical resection, radiotherapy and chemotherapy, the median survival of malignant glioma patients remain poor

  • We studied the effect of celecoxib in human glioblastoma cells with various p53 status; U87MG cells with high and low levels of p53 [by both genetic and pharmaceutical intervention], LN229 and U373MG cells

  • U87MG cells were pre-treated with PFT (25 μM) for 30 minutes prior to celecoxib treatment

Read more

Summary

Introduction

Despite conventional therapy of surgical resection, radiotherapy and chemotherapy, the median survival of malignant glioma patients remain poor. Targeting COX-2 with selective COX-2 inhibitors (NS-398, SC-236 and celecoxib) has proven effective to reduce human glioblastoma cell viability in vitro [4,6,7,8,9] and in rodent models [6,9,10,11]. Celecoxib is the only selective COX-2 inhibitor approved by the FDA for adjuvant treatment of patients with familial adenomatous polyposis. Selective cyclooxygenase (COX)-2 inhibitors elicit anti-proliferative responses in various tumours, the underlying anti-tumour mechanisms are unclear. The role of p53 mutation in the anti-tumour responses of the selective COX-2 inhibitor celecoxib in human glioblastoma cells is unknown. We investigated whether the anti-glioblastoma responses of celecoxib were p53dependent, and whether celecoxib induced DNA damage leading to p53-dependent G1 cell cycle arrest, followed by autophagy or apoptosis

Methods
Results
Discussion
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