Synuclein γ compromises spindle assembly checkpoint and renders resistance to antimicrotubule drugs

S Miao, K Wu, B Zhang, Z Weng, M Zhu, Y Lu… - Molecular cancer …, 2014 - AACR
S Miao, K Wu, B Zhang, Z Weng, M Zhu, Y Lu, R Krishna, YE Shi
Molecular cancer therapeutics, 2014AACR
Defects in the spindle assembly checkpoint (SAC) have been proposed to contribute to the
chromosomal instability in human cancers. One of the major mechanisms underlying
antimicrotubule drug (AMD) resistance involves acquired inactivation of SAC. Synuclein γ
(SNCG), previously identified as a breast cancer–specific gene, is highly expressed in
malignant cancer cells but not in normal epithelium. Here, we show that SNCG is sufficient to
induce resistance to AMD-caused apoptosis in breast cancer cells and cancer xenografts …
Abstract
Defects in the spindle assembly checkpoint (SAC) have been proposed to contribute to the chromosomal instability in human cancers. One of the major mechanisms underlying antimicrotubule drug (AMD) resistance involves acquired inactivation of SAC. Synuclein γ (SNCG), previously identified as a breast cancer–specific gene, is highly expressed in malignant cancer cells but not in normal epithelium. Here, we show that SNCG is sufficient to induce resistance to AMD-caused apoptosis in breast cancer cells and cancer xenografts. SNCG binds to spindle checkpoint kinase BubR1 and inhibits its kinase activity. Specifically, the C-terminal (Gln106-Asp127) of SNCG binds to the N-terminal TPR (tetratricopeptidelike folds) motif of BubR1. SNCG–BubR1 interaction induces a structure change of BubR1, attenuates its interaction with other key checkpoint proteins of Cdc20, and thus compromises SAC function. SNCG expression in breast cancers from patients with a neoadjuvant clinical trial showed that SNCG-positive tumors are resistant to chemotherapy-induced apoptosis. These data show that SNCG renders AMD resistance by inhibiting BubR1 activity and attenuating SAC function. Mol Cancer Ther; 13(3); 699–713. ©2014 AACR.
AACR