Your browser doesn't support javascript.
loading
CHD4 R975H mutant activates tumorigenic pathways and promotes stemness and M2-like macrophage polarization in endometrial cancer.
Zhang, Qinglin; Zhu, Fengzhi; Tong, Yin; Shi, Diwen; Zhang, Jiangwen.
Afiliação
  • Zhang Q; School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China.
  • Zhu F; College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
  • Tong Y; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
  • Shi D; School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China.
  • Zhang J; School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China. jzhang1@hku.hk.
Sci Rep ; 14(1): 18617, 2024 08 10.
Article em En | MEDLINE | ID: mdl-39127769
ABSTRACT
Endometrial cancer (EC), one of the most prevalent carcinomas in females, is associated with increasing mortality. We identified the CHD4 R975H mutation as a high-frequency occurrence in EC patients through a comprehensive survey of EC databases. Computational predictions suggest that this mutation profoundly impacts the structural and functional integrity of CHD4. Functional assays revealed that the CHD4 R975H mutation enhances EC cell invasion, proliferation, and colony formation, promoting a cancer stem cell (CSC)-like phenotype. RNA-seq analysis of cells expressing CHD4 R975H mutant revealed a transcriptomic landscape marked by the activation of several cancer-promoting signaling pathways, including TNF-α signaling via NF-κB, KRAS, P53, mTOR, TGF-ß, EGFR, Myc and growth factor signaling. Validation assays confirmed the activation of these pathways, further demonstrating that CHD4 R975H mutation induces stemness in EC cells and M2-like polarization of tumor-associated macrophages (TAMs). Our study elucidated the oncogenic role of CHD4 R975H mutation, highlighting its dual impact on facilitating cancer stemness and transforming TAMs into an immunosuppressive subtype. These findings contribute valuable insights into the molecular mechanisms driving EC progression and open avenues for targeted therapeutic interventions.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Transdução de Sinais / Neoplasias do Endométrio / Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Transdução de Sinais / Neoplasias do Endométrio / Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2024 Tipo de documento: Article