1. Academic Validation
  2. Discovery of a novel inhibitor of kinesin-like protein KIFC1

Discovery of a novel inhibitor of kinesin-like protein KIFC1

  • Biochem J. 2016 Apr 15;473(8):1027-35. doi: 10.1042/BJ20150992.
Wei Zhang 1 Ling Zhai 2 Yimin Wang 2 Rebecca J Boohaker 2 Wenyan Lu 2 Vandana V Gupta 2 Indira Padmalayam 2 Robert J Bostwick 2 E Lucile White 2 Larry J Ross 2 Joseph Maddry 2 Subramaniam Ananthan 2 Corinne E Augelli-Szafran 2 Mark J Suto 2 Bo Xu 2 Rongbao Li 2 Yonghe Li 2
Affiliations

Affiliations

  • 1 Division of Drug Discovery, Southern Research Institute, Birmingham, AL 35205, U.S.A. [email protected].
  • 2 Division of Drug Discovery, Southern Research Institute, Birmingham, AL 35205, U.S.A.
Abstract

Historically, drugs used in the treatment of cancers also tend to cause damage to healthy cells while affecting Cancer cells. Therefore, the identification of novel agents that act specifically against Cancer cells remains a high priority in the search for new therapies. In contrast with normal cells, most Cancer cells contain multiple centrosomes which are associated with genome instability and tumorigenesis. Cancer cells can avoid multipolar mitosis, which can cause cell death, by clustering the extra centrosomes into two spindle poles, thereby enabling bipolar division. Kinesin-like protein KIFC1 plays a critical role in centrosome clustering in Cancer cells, but is not essential for normal cells. Therefore, targeting KIFC1 may provide novel insight into selective killing of Cancer cells. In the present study, we identified a small-molecule KIFC1 inhibitor, SR31527, which inhibited microtubule (MT)-stimulated KIFC1 ATPase activity with an IC50 value of 6.6 μM. By using bio layer interferometry technology, we further demonstrated that SR31527 bound directly to KIFC1 with high affinity (Kd=25.4 nM). Our results from computational modelling and saturation-transfer difference (STD)-NMR experiments suggest that SR31527 bound to a novel allosteric site of KIFC1 that appears suitable for developing selective inhibitors of KIFC1. Importantly, SR31527 prevented bipolar clustering of extra centrosomes in triple negative breast Cancer (TNBC) cells and significantly reduced TNBC cell colony formation and viability, but was less toxic to normal fibroblasts. Therefore, SR31527 provides a valuable tool for studying the biological function of KIFC1 and serves as a potential lead for the development of novel therapeutic agents for breast Cancer treatment.

Keywords

allosteric regulation; breast cancer; inhibitor; kinesin; mitosis.

Figures
Products