Your browser doesn't support javascript.
loading
C-terminal kinesin motor KIFC1 participates in facilitating proper cell division of human seminoma.
Xiao, Yu-Xi; Shen, Hao-Qing; She, Zhen-Yu; Sheng, Li; Chen, Qian-Qian; Chu, Yu-Lan; Tan, Fu-Qing; Yang, Wan-Xi.
Afiliación
  • Xiao YX; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
  • Shen HQ; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
  • She ZY; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
  • Sheng L; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
  • Chen QQ; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
  • Chu YL; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
  • Tan FQ; The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
  • Yang WX; The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, China.
Oncotarget ; 8(37): 61373-61384, 2017 Sep 22.
Article en En | MEDLINE | ID: mdl-28977870
ABSTRACT
C-terminus kinesin motor KIFC1 is known for centrosome clustering in cancer cells with supernumerary centrosomes. KIFC1 crosslinks and glides on microtubules (MT) to assist normal bipolar spindle formation to avoid multi-polar cell division, which might be fatal. Testis cancer is the most common human cancer among young men. However, the gene expression profiles of testis cancer is still not complete and the expression of the C-terminus kinesin motor KIFC1 in testis cancer has not yet been examined. We found that KIFC1 is enriched in seminoma tissues in both mRNA level and protein level, and is specifically enriched in the cells that divide actively. Cell experiments showed that KIFC1 may be essential in cell division, but not essential in metastasis. Based on subcellular immuno-florescent staining results, we also described the localization of KIFC1 during cell cycle. By expressing ΔC-FLAG peptide in the cells, we found that the tail domain of KIFC1 might be essential for the dynamic disassociation of KIFC1, and the motor domain of KIFC1 might be essential for the degradation of KIFC1. Our work provides a new perspective for seminoma research.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Oncotarget Año: 2017 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Oncotarget Año: 2017 Tipo del documento: Article País de afiliación: China
...