Your browser doesn't support javascript.
loading
Nuclear factor I-C regulates E-cadherin via control of KLF4 in breast cancer.
Lee, Hye-Kyung; Lee, Dong-Seol; Park, Joo-Cheol.
Afiliación
  • Lee HK; Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehagro, Chongro-gu, Seoul, 110-749, South Korea. h35071221@snu.ac.kr.
  • Lee DS; Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehagro, Chongro-gu, Seoul, 110-749, South Korea. iceburge@snu.ac.kr.
  • Park JC; Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, 101 Daehagro, Chongro-gu, Seoul, 110-749, South Korea. jcapark@snu.ac.kr.
BMC Cancer ; 15: 113, 2015 Mar 10.
Article en En | MEDLINE | ID: mdl-25879941
ABSTRACT

BACKGROUND:

Progression to metastasis is the leading cause of most cancer-related mortality; however, much remains to be understood about what facilitates the spread of tumor cells. In the present study, we describe a novel pathway in breast cancer that regulates epithelial-to-mesenchymal transition (EMT), motility, and invasiveness.

METHODS:

We examined nuclear factor I-C (NFI-C) expression in MCF10A human breast epithelial cells, MCF7 non-invasive breast cancer cells, and MDA-MB231 invasive breast cancer cells by real-time PCR and western blotting. To investigate the loss- and gain-function of NFI-C, we determined whether NFI-C regulated KLF4 expression by real-time PCR, western blotting, and promoter assay. To understand the biological functions of NFI-C, we observed cell invasion, migration, adhesion in human tumor cells by transwell assay, wound healing assay, quantitative RT-PCR, cell adhesion assay, western blotting, and immunohistochemistry.

RESULTS:

We identified the downstream factors of NFI-C, such as KLF4 and E-cadherin, which play roles in EMT. NFI-C is expressed in normal mammary gland or noninvasive breast cancer cells with epithelial characteristics. NFI-C overexpression induced expression of KLF4 and E-cadherin, but not Slug, in breast cancer cells. NFI-C bound directly to the KLF4 promoter and stimulated KLF4 transcriptional activity, thereby regulating E-cadherin expression during tumorigenesis. Cells overexpressing NFI-C maintained their epithelial differentiation status, which could drive mesenchymal-epithelial transition (MET) via the NFI-C-KLF4-E-cadherin axis in breast cancer cells. Consequently, NFI-C suppressed EMT, migration, and invasion in breast cancer cells.

CONCLUSIONS:

Our study reveals a novel signaling pathway that is important during breast cancer tumorigenesis the NFI-C-KLF4-E-cadherin pathway. The results indicate the important role of NFI-C in regulating KLF4 during tumorigenesis.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Mama / Cadherinas / Factores de Transcripción de Tipo Kruppel / Factores de Transcripción NFI Tipo de estudio: Prognostic_studies Límite: Female / Humans Idioma: En Revista: BMC Cancer Asunto de la revista: NEOPLASIAS Año: 2015 Tipo del documento: Article País de afiliación: Corea del Sur

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Mama / Cadherinas / Factores de Transcripción de Tipo Kruppel / Factores de Transcripción NFI Tipo de estudio: Prognostic_studies Límite: Female / Humans Idioma: En Revista: BMC Cancer Asunto de la revista: NEOPLASIAS Año: 2015 Tipo del documento: Article País de afiliación: Corea del Sur