Your browser doesn't support javascript.
loading
HDAC2- and EZH2-Mediated Histone Modifications Induce PDK1 Expression through miR-148a Downregulation in Breast Cancer Progression and Adriamycin Resistance.
Xie, Yunxia; Shi, Zhumei; Qian, Yingchen; Jiang, Chengfei; Liu, Wenjing; Liu, Bingjie; Jiang, Binghua.
Afiliación
  • Xie Y; Academy of Medical Science, School of Basic Medical Sciences, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
  • Shi Z; Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China.
  • Qian Y; Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China.
  • Jiang C; Department of Pathology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China.
  • Liu W; Academy of Medical Science, School of Basic Medical Sciences, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
  • Liu B; Academy of Medical Science, School of Basic Medical Sciences, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
  • Jiang B; Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
Cancers (Basel) ; 14(15)2022 Jul 23.
Article en En | MEDLINE | ID: mdl-35892859
ABSTRACT

BACKGROUND:

Breast cancer has one of highest morbidity and mortality rates for women. Abnormalities regarding epigenetics modification and pyruvate dehydrogenase kinase 1 (PDK1)-induced unusual metabolism contribute to breast cancer progression and chemotherapy resistance. However, the role and mechanism of epigenetic change in regulating PDK1 in breast cancer remains to be elucidated.

METHODS:

Gene set enrichment analysis (GSEA) and Pearson's correlation analysis were performed to analyze the relationship between histone deacetylase 2 (HDAC2), enhancer of zeste homologue 2 (EZH2), and PDK1 in database and human breast cancer tissues. Dual luciferase reporters were used to test the regulation between PDK1 and miR-148a. HDAC2 and EZH2 were found to regulate miR-148a expression through Western blotting assays, qRT-PCR and co-immunoprecipitation assays. The effects of PDK1 and miR-148a in breast cancer were investigated by immunofluorescence (IF) assay, Transwell assay and flow cytometry assay. The roles of miR-148a/PDK1 in tumor growth were investigated in vivo.

RESULTS:

We found that PDK1 expression was upregulated by epigenetic alterations mediated by HDAC2 and EZH2. At the post-transcriptional level, PDK1 was a new direct target of miR-148a and was upregulated in breast cancer cells due to miR-148a suppression. PDK1 overexpression partly reversed the biological function of miR-148a-including miR-148a's ability to increase cell sensitivity to Adriamycin (ADR) treatment-inhibiting cell glycolysis, invasion and epithelial-mesenchymal transition (EMT), and inducing apoptosis and repressing tumor growth. Furthermore, we identified a novel mechanism DNMT1 directly bound to EZH2 and recruited EZH2 and HDAC2 complexes to the promoter region of miR-148a, leading to miR-148a downregulation. In breast cancer tissues, HDAC2 and EZH2 protein expression levels also were inversely correlated with levels of miR-148a expression.

CONCLUSION:

Our study found a new regulatory mechanism in which EZH2 and HDAC2 mediate PDK1 upregulation by silencing miR-148a expression to regulate cancer development and Adriamycin resistance. These new findings suggest that the HDAC2/EZH2/miR-148a/PDK1 axis is a novel mechanism for regulating cancer development and is a potentially promising target for therapeutic options in the future.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Cancers (Basel) Año: 2022 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Cancers (Basel) Año: 2022 Tipo del documento: Article País de afiliación: China