Your browser doesn't support javascript.
loading
C9orf142 transcriptionally activates MTBP to drive progression and resistance to CDK4/6 inhibitor in triple-negative breast cancer.
Liao, Li; Deng, Ling; Zhang, Yin-Ling; Yang, Shao-Ying; Andriani, Lisa; Hu, Shu-Yuan; Zhang, Fang-Lin; Shao, Zhi-Min; Li, Da-Qiang.
Afiliación
  • Liao L; Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
  • Deng L; Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.
  • Zhang YL; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
  • Yang SY; Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
  • Andriani L; Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.
  • Hu SY; Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
  • Zhang FL; Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.
  • Shao ZM; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
  • Li DQ; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.
Clin Transl Med ; 13(11): e1480, 2023 11.
Article en En | MEDLINE | ID: mdl-38009308
ABSTRACT

BACKGROUND:

Triple-negative breast cancer (TNBC) presents the most challenging subtype of all breast cancers because of its aggressive clinical phenotypes and absence of viable therapy targets. In order to identify effective molecular targets for treating patients with TNBC, we conducted an integration analysis of our recently published TNBC dataset of quantitative proteomics and RNA-Sequencing, and found the abnormal upregulation of chromosome 9 open reading frame 142 (C9orf142) in TNBC. However, the functional roles of C9orf142 in TNBC are unclear.

METHODS:

In vitro and in vivo functional experiments were performed to assess potential roles of C9orf142 in TNBC. Immunoblotting, real-time quantitative polymerase chain reaction (RT-qPCR), and immunofluorescent staining were used to investigate the expression levels of C9orf142 and its downstream molecules. The molecular mechanisms underlying C9orf142-regulated mouse double minute 2 (MDM2)-binding protein (MTBP) were determined by chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays.

RESULTS:

In TNBC tissues and metastatic lymph nodes, we observed that C9orf142 exhibited abnormal up-regulation, and its elevated expression was indicative of unfavorable prognosis for TNBC patients. Both in vitro and in vivo functional experiments demonstrated that C9orf142 accelerated TNBC growth and metastasis. Further mechanism exploration revealed that C9orf142 transcriptionally activated MTBP, thereby regulating its downstream MDM2/p53/p21 signaling axis and the transition of cell cycle from G1 to S phase. Functional rescue experiment demonstrated that knockdown of MTBP attenuated C9orf142-mediated tumour growth and metastasis. Furthermore, depletion of C9orf142 remarkably increased the responsiveness of TNBC cells to CDK4/6 inhibitor abemaciclib.

CONCLUSIONS:

Together, these findings unveil a previously unrecognized effect of C9orf142 in TNBC progression and responsiveness to CDK4/6 inhibitor, and emphasize C9orf142 as a promising intervention target for TNBC treatment.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Mama Triple Negativas Límite: Animals / Humans Idioma: En Revista: Clin Transl Med Año: 2023 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Mama Triple Negativas Límite: Animals / Humans Idioma: En Revista: Clin Transl Med Año: 2023 Tipo del documento: Article País de afiliación: China