Abstract

Highly expressed in cancer protein 1 (Hec1) is a subunit of the kinetochore (KT)-associated Ndc80 complex, which ensures proper segregation of sister chromatids at mitosis by mediating the interaction between KTs and microtubules (MTs). HEC1 mRNA and protein are highly expressed in many malignancies as part of a signature of chromosome instability. These properties render Hec1 a promising molecular target for developing therapeutic drugs that exert their anticancer activities by producing massive chromosome aneuploidy. A virtual screening study aimed at identifying small molecules able to bind at the Hec1–MT interaction domain identified one positive hit compound and two analogs of the hit with high cytotoxic, pro-apoptotic and anti-mitotic activities. The most cytotoxic analog (SM15) was shown to produce chromosome segregation defects in cancer cells by inhibiting the correction of erroneous KT–MT interactions. Live cell imaging of treated cells demonstrated that mitotic arrest and segregation abnormalities lead to cell death through mitotic catastrophe and that cell death occurred also from interphase. Importantly, SM15 was shown to be more effective in inducing apoptotic cell death in cancer cells as compared to normal ones and effectively reduced tumor growth in a mouse xenograft model. Mechanistically, cold-induced MT depolymerization experiments demonstrated a hyper-stabilization of both mitotic and interphase MTs. Molecular dynamics simulations corroborate this finding by showing that SM15 can bind the MT surface independently from Hec1 and acts as a stabilizer of both MTs and KT–MT interactions. Overall, our studies represent a clear proof of principle that MT-Hec1-interacting compounds may represent novel powerful anticancer agents.

Highlights

  • Aneuploidy and chromosome instability are strongly involved in tumorigenesis

  • To gain insights into the molecular mechanism mediating the interaction of SM15-treated cells (SM15) with its targets, we investigated the modification exerted by SM15 on MTs and KT–MT attachments at the molecular level by carrying out a molecular dynamics (MD) simulation of the tubulin dimer–Hec[1] complex in the presence of SM15

  • These findings are in line with the surface plasmon resonance (SPR) results, in which SM15 was able to bind directly and with high affinity to MTs and to tubulin

Read more

Summary

Introduction

Aneuploidy and chromosome instability are strongly involved in tumorigenesis. Studies on the cancer susceptibility of mouse models heterozygous for genes controlling chromosome segregation suggest that aneuploidy may act as a tumor suppressive mechanism, depending on the tissue analyzed and its intrinsic chromosome instability.[1,2] This evidence suggests that excessive chromosome mis-segregation may compromise genome stability and be incompatible with cell viability. Partial downregulation of mitotic checkpoint proteins in tumor cell lines produced cancer cell death associated with severe chromosome mis-segregation.[3] overexpression of cyclin E inducing multipolar divisions and numerous chromosome segregation errors was found to have a significant antitumorigenic effect in a mouse model for lung cancer.[4] the idea of promoting cell death by inducing massive chromosome mis-segregation at mitotic division was proposed as a therapeutic strategy to selectively eliminate actively proliferating tumor cells.[5]

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