Your browser doesn't support javascript.
loading
A novel role for KIFC1-MYH9 interaction in triple-negative breast cancer aggressiveness and racial disparity.
Garlapati, Chakravarthy; Joshi, Shriya; Yang, Chunhua; Chandrashekar, Darshan Shimoga; Rida, Padmashree; Aneja, Ritu.
Afiliación
  • Garlapati C; Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
  • Joshi S; Alkermes Inc, Waltham, MA, 02451, USA.
  • Yang C; Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
  • Chandrashekar DS; Alkermes Inc, Waltham, MA, 02451, USA.
  • Rida P; Small molecule drug discovery, Bristol Myers Squibb, Cambridge, MA, 02141, USA.
  • Aneja R; Institute of Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
Cell Commun Signal ; 22(1): 312, 2024 Jun 06.
Article en En | MEDLINE | ID: mdl-38902769
ABSTRACT
African American (AA) women are twice as likely to develop triple-negative breast cancer (TNBC) as women of European descent. Additionally, AA women with TNBC present a much more aggressive disease course than their European American (EA) counterparts. Thus, there is an unmet clinical need to identify race-specific biomarkers and improve survival outcomes in AA patients with TNBC. The minus-end directed microtubule motor protein kinesin family member C1 (KIFC1) promotes centrosome clustering and chromosomal instability and is often overexpressed in TNBC. Previous findings suggest that KIFC1 plays a role in cell proliferation and migration in TNBC cells from AAs and that the levels of nuclear KIFC1 (nKIFC1) are particularly high in AA patients with TNBC. The nuclear localization of KIFC1 in interphase may underlie its previously unrecognized race-specific association. In this study, we found that in TNBC cells derived from AAs, nKIFC1 interacted with the tumor suppressor myosin heavy chain 9 (MYH9) over EA cells. Treatment of AA TNBC cells with commercial inhibitors of KIFC1 and MYH9 disrupted the interaction between KIFC1 and MYH9. To characterize the racial differences in the KIFC1-MYH9-MYC axis in TNBC, we established homozygous KIFC1 knockout (KO) TNBC cell lines. KIFC1 KO significantly inhibited proliferation, migration, and invasion in AA TNBC cells but not in EA TNBC cells. RNA sequencing analysis showed significant downregulation of genes involved in cell migration, invasion, and metastasis upon KIFC1 KO in TNBC cell lines from AAs compared to those from EAs. These data indicate that mechanistically, the role of nKIFC1 in driving TNBC progression and metastasis is stronger in AA patients than in EA patients, and that KIFC1 may be a critical therapeutic target for AA patients with TNBC.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Cinesinas / Cadenas Pesadas de Miosina / Neoplasias de la Mama Triple Negativas Límite: Female / Humans Idioma: En Revista: Cell Commun Signal Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Cinesinas / Cadenas Pesadas de Miosina / Neoplasias de la Mama Triple Negativas Límite: Female / Humans Idioma: En Revista: Cell Commun Signal Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos
...