Your browser doesn't support javascript.
loading
In-depth Profiling and Quantification of the Lysine Acetylome in Hepatocellular Carcinoma with a Trapped Ion Mobility Mass Spectrometer.
Xu, Jia; Guan, Xinyu; Jia, Xiaodong; Li, Hongyan; Chen, Ruibing; Lu, Yinying.
  • Xu J; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
  • Guan X; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
  • Jia X; Department of Hepatology, Comprehensive Liver Cancer Center, The Fifth Medical Center of General Hospital of PLA, Beijing, China.
  • Li H; General surgery Department, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China. Electronic address: hongyanli@xwhosp.org.
  • Chen R; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China. Electronic address: rbchen@tju.edu.cn.
  • Lu Y; Guangdong Key Laboratory of Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China. Electronic address: luyinying1973@163.com.
Mol Cell Proteomics ; 21(8): 100255, 2022 08.
Article en En | MEDLINE | ID: mdl-35688384
ABSTRACT
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death worldwide with limited therapeutic options. Comprehensive investigation of protein posttranslational modifications in HCC is still limited. Lysine acetylation is one of the most common types of posttranslational modification involved in many cellular processes and plays crucial roles in the regulation of cancer. In this study, we analyzed the proteome and K-acetylome in eight pairs of HCC tumors and normal adjacent tissues using a timsTOF Pro instrument. As a result, we identified 9219 K-acetylation sites in 2625 proteins, of which 1003 sites exhibited differential acetylation levels between tumors and normal adjacent tissues. Interestingly, many novel tumor-specific K-acetylation sites were characterized, for example, filamin A (K865), filamin B (K697), and cofilin (K19), suggesting altered activities of these cytoskeleton-modulating molecules, which may contribute to tumor metastasis. In addition, we observed an overall suppression of protein K-acetylation in HCC tumors, especially for enzymes from various metabolic pathways, for example, glycolysis, tricarboxylic acid cycle, and fatty acid metabolism. Moreover, the expression of deacetylase sirtuin 2 (SIRT2) was upregulated in HCC tumors, and its role of deacetylation in HCC cells was further explored by examining the impact of SIRT2 overexpression on the proteome and K-acetylome in Huh7 HCC cells. SIRT2 overexpression reduced K-acetylation of proteins involved in a wide range of cellular processes, including energy metabolism. Furthermore, cellular assays showed that overexpression of SIRT2 in HCC cells inhibited both glycolysis and oxidative phosphorylation. Taken together, our findings provide valuable information to better understand the roles of K-acetylation in HCC and to treat this disease by correcting the aberrant acetylation patterns.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Carcinoma Hepatocelular / Neoplasias Hepáticas Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Año: 2022 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Carcinoma Hepatocelular / Neoplasias Hepáticas Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Año: 2022 Tipo del documento: Article