Abstract

SummaryIntroduction Differentiation therapy is a promising strategy for cancer treatment. The translationally controlled tumor protein (TCTP) is an encouraging target in this context. By now, this field of research is still at its infancy, which motivated us to perform a large-scale screening for the identification of novel ligands of TCTP. We studied the binding mode and the effect of TCTP blockade on the cell cycle in different cancer cell lines. Methods Based on the ZINC-database, we performed virtual screening of 2,556,750 compounds to analyze the binding of small molecules to TCTP. The in silico results were confirmed by microscale thermophoresis. The effect of the new ligand molecules was investigated on cancer cell survival, flow cytometric cell cycle analysis and protein expression by Western blotting and co-immunoprecipitation in MOLT-4, MDA-MB-231, SK-OV-3 and MCF-7 cells. Results Large-scale virtual screening by PyRx combined with molecular docking by AutoDock4 revealed five candidate compounds. By microscale thermophoresis, ZINC10157406 (6-(4-fluorophenyl)-2-[(8-methoxy-4-methyl-2-quinazolinyl)amino]-4(3H)-pyrimidinone) was identified as TCTP ligand with a KD of 0.87 ± 0.38. ZINC10157406 revealed growth inhibitory effects and caused G0/G1 cell cycle arrest in MOLT-4, SK-OV-3 and MCF-7 cells. ZINC10157406 (2 × IC50) downregulated TCTP expression by 86.70 ± 0.44% and upregulated p53 expression by 177.60 ± 12.46%. We validated ZINC10157406 binding to the p53 interaction site of TCTP and replacing p53 by co-immunoprecipitation. Discussion ZINC10157406 was identified as potent ligand of TCTP by in silico and in vitro methods. The compound bound to TCTP with a considerably higher affinity compared to artesunate as known TCTP inhibitor. We were able to demonstrate the effect of TCTP blockade at the p53 binding site, i.e. expression of TCTP decreased, whereas p53 expression increased. This effect was accompanied by a dose-dependent decrease of CDK2, CDK4, CDK, cyclin D1 and cyclin D3 causing a G0/G1 cell cycle arrest in MOLT-4, SK-OV-3 and MCF-7 cells. Our findings are supposed to stimulate further research on TCTP-specific small molecules for differentiation therapy in oncology.

Highlights

  • Materials and methodsDue to its high involvement in various biological functions, the translationally controlled tumor protein (TCTP) gained increasing interest since its discovery in 1983 in mouse tumor cells [1, 2]

  • We chose a computational approach as efficient way to evaluate the binding of potential chemical binders to TCTP

  • We generated our own chemical library derived from the ZINC database (Fig. 1)

Read more

Summary

Introduction

Materials and methodsDue to its high involvement in various biological functions, the translationally controlled tumor protein (TCTP) gained increasing interest since its discovery in 1983 in mouse tumor cells [1, 2]. TCTP is an evolutionary highly conserved protein with functions in cell growth, protein synthesis, cytoskeleton, immune response, development, malignant transformation, tumor reversion, induction of pluripotent stem cells and apoptosis It interacts with a large number of proteins [3]. Modern cancer therapy is still dependent on classical chemotherapy with a significant lack in tumor selectivity leading to non-tolerable side effects. Because of these side effects, sufficiently high doses cannot be applied to patients to eradicate all tumor cells in the body. Differentiation therapy represents an alternative concept pledging higher tumor selectivity and lower side effects It aims at restoring cellular differentiation and re-gaining cells into normal cellular programs [13]. We performed a focused molecular in silico screening of the ZINC database comprising of 2,556,750 chemical compounds and identified

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