Your browser doesn't support javascript.
loading
Metabolic self-feeding in HBV-associated hepatocarcinoma centered on feedback between circulation lipids and the cellular MAPK/mTOR axis.
Zhu, Ying; Zhao, Yingke; Ning, Zhouyu; Deng, Yong; Li, Bing; Sun, Yun; Meng, Zhiqiang.
Afiliação
  • Zhu Y; Minimally invasive therapy center, Shanghai Cancer Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
  • Zhao Y; Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
  • Ning Z; Minimally invasive therapy center, Shanghai Cancer Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
  • Deng Y; Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China.
  • Li B; Minimally invasive therapy center, Shanghai Cancer Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
  • Sun Y; Department of Research and Development, Department of Nuclear Medicine, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, 201321, China.
  • Meng Z; Shanghai Key Laboratory of radiation oncology (20dz2261000), Shanghai, 201321, China.
Cell Commun Signal ; 22(1): 280, 2024 May 21.
Article em En | MEDLINE | ID: mdl-38773448
ABSTRACT

INTRODUCTION:

Hepatitis B Virus (HBV) is widely recognized as a "metabolic virus" that disrupts hepatic metabolic homeostasis, rendering it one of the foremost risk factors for hepatocellular carcinoma (HCC). Except for antiviral therapy, the fundamental principles underlying HBV- and HBV+ HCC have remained unchanged, limiting HCC treatment options.

OBJECTIVES:

In this study, we aim to identify the distinctive metabolic profile of HBV-associated HCC, with the promise of identifying novel metabolic targets that confer survival advantages and ultimately impede cancer progression.

METHODS:

We employed a comprehensive methodology to evaluate metabolic alterations systematically. Initially, we analyzed transcriptomic and proteomic data obtained from a public database, subsequently validating these findings within our test cohort at both the proteomic and transcriptomic levels. Additionally, we conducted a comprehensive analysis of tissue metabolomics profiles, lipidomics, and the activity of the MAPK and AKT signaling pathway to corroborate the abovementioned changes.

RESULTS:

Our multi-omics approach revealed distinct metabolic dysfunctions associated with HBV-associated HCC. Specifically, we observed upregulated steroid hormone biosynthesis, primary bile acid metabolism, and sphingolipid metabolism in HBV-associated HCC patients' serum. Notably, metabolites involved in primary bile acid and sphingolipids can activate the MAPK/mTOR pathway. Tissue metabolomics and lipidomics analyses further validated the serum metabolic alterations, particularly alterations in lipid composition and accumulation of unsaturated fatty acids.

CONCLUSION:

Our findings emphasize the pivotal role of HBV in HCC metabolism, elucidating the activation of a unique MAPK/mTOR signaling axis by primary bile acids and sphingolipids. Moreover, the hyperactive MAPK/mTOR signaling axis transduction leads to significant reprogramming in lipid metabolism within HCC cells, further triggering the activation of the MAPK/mTOR pathway in turn, thereby establishing a self-feeding circle driven by primary bile acids and sphingolipids.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Vírus da Hepatite B / Carcinoma Hepatocelular / Serina-Treonina Quinases TOR / Neoplasias Hepáticas Limite: Female / Humans / Male / Middle aged Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Vírus da Hepatite B / Carcinoma Hepatocelular / Serina-Treonina Quinases TOR / Neoplasias Hepáticas Limite: Female / Humans / Male / Middle aged Idioma: En Ano de publicação: 2024 Tipo de documento: Article