Abstract

BackgroundLycorine has been revealed to inhibit the development of many kinds of malignant tumors, including glioblastoma multiforme (GBM). Although compelling evidences demonstrated Lycorine’s inhibition on cancers through some peripheral mechanism, in-depth mechanism studies of Lycotine’s anti-GBM effects still call for further exploration. Epidermal Growth Factor Receptor (EGFR) gene amplification and mutations are the most common oncogenic events in GBM. Targeting EGFR by small molecular inhibitors is a rational strategy for GBM treatment.MethodsThe molecular docking modeling and in vitro EGFR kinase activity system were employed to identify the potential inhibitory effects of Lycorine on EGFR. And the Biacore assay was used to confirm the direct binding status between Lycorine and the intracellular EGFR (696–1022) domain. In vitro assays were conducted to test the suppression of Lycorine on the biological behavior of GBM cells. By RNA interference, EGFR expression was reduced then cells underwent proliferation assay to investigate whether Lycorine’s inhibition on GBM cells was EGFR-dependent or not. RT-PCR and western blotting analysis were carried out to investigate the underlined molecular mechanism that Lycorine exerted on EGFR itself and EGFR signaling pathway. Three different xenograft models (an U251-luc intracranially orthotopic transplantation model, an EGFR stably knockdown U251 subcutaneous xenograft model and a patient-derived xenograft model) were performed to verify Lycorine’s therapeutic potential on GBM in vivo.ResultsWe identified a novel small natural molecule Lycorine binding to the intracellular EGFR (696–1022) domain as an inhibitor of EGFR. Lycorine decreased GBM cell proliferation, migration and colony formation by inducing cell apoptosis in an EGFR-mediated manner. Furthermore, Lycorine inhibited the xenograft tumor growths in three animal models in vivo. Besides, Lycorine impaired the phosphorylation of EGFR, AKT, which were mechanistically associated with expression alteration of a series of cell survival and death regulators and metastasis-related MMP9 protein.ConclusionsOur findings identify Lycorine directly interacts with EGFR and inhibits EGFR activation. The most significant result is that Lycorine displays satisfactory therapeutic effect in our patient-derived GBM tumor xenograft, thus supporting the conclusion that Lycorine may be considered as a promising candidate in clinical therapy for GBM.

Highlights

  • Lycorine has been revealed to inhibit the development of many kinds of malignant tumors, including glioblastoma multiforme (GBM)

  • Investigations of Lycorine’s interaction with Epidermal Growth Factor Receptor (EGFR) have not been described in previous literature. We present in this current study that Lycorine inhibits proliferation and migration of various GBM cell lines,including cells holding wild type EGFR amplification and EGFRvIII, and induces cell apoptosis and cell death

  • Molecular docking modeling assay The X-ray crystal structure of EGFR was obtained from the Protein data bank ((PDB ID: 5FED, EGFR kinase domain in complex with a covalent aminobenzimidazole inhibitor) website

Read more

Summary

Introduction

Lycorine has been revealed to inhibit the development of many kinds of malignant tumors, including glioblastoma multiforme (GBM). Epidermal Growth Factor Receptor (EGFR) gene amplification and mutations are the most common oncogenic events in GBM. EGFR activation leads to autophosphorylation of several key tyrosine residues triggering several intracellular downstream signaling pathways including the Ras/Raf/MEK/ ERK pathway, the PLCγ -PKC pathway and the PI3K/ AKT pathway, resulting in cell proliferation, motility and survival [13]. Within such a large proportion of EGFR genomic alterations, approximately 20–40% of them harbor EGFR variant III (EGFRvIII) mutant, which contains a deletion of exons 2–7 in the extracellular ligand-binding domain [14, 15]. EGFRvIII induces the receptor tyrosine kinase activation in both a cell autonomous and nonautonomous manner, results in a ligand-independent mutant and shows constitutive activation in the absence of ligand to activate the tumor-promoting signaling pathways [16]

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.