Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Background & Aims: Hepatocellular carcinoma (HCC) frequently involves metabolic reprogramming, which promotes oncogenesis and metastasis. However, the underlying molecular mechanisms remain insufficiently explored. In this study, we aim to investigate the metabolic abnormalities in c-Myc-driven HCC development and their potential therapeutic implications.

Methods: RNA sequencing and metabolomics were performed on HCC and adjacent tissues in a murine HCC model established by hydrodynamic tail-vein injection of c-Myc and sgTrp53/Cas9 plasmids. Key catalytic enzyme gene knockout was used to assess tumor formation and murine survival. Gene expression was analyzed using quantitative polymerase chain reaction, immunohistochemistry, and Western blot. Chromatin immunoprecipitation followed by quantitative polymerase chain reaction and luciferase assays verified c-Myc regulation.

Results: RNA sequencing data revealed that the hexosamine biosynthetic pathway was significantly activated in c-Myc-driven HCC. The rate-limiting enzyme GFPT1 (rather than GFPT2) was up-regulated in the first step of this pathway. Knocking out GFPT1 reduces tumor growth and prolongs murine survival. Human specimens showed that GFPT1 was overexpressed in HCC tissues and was associated with advanced Edmondson-Steiner grades and short patient survival. Further luciferase reporter assays confirmed that c-Myc binds directly to the promoter region of GFPT1 and activates its transcription. Subsequent examination of the downstream pathways of the hexosamine biosynthetic pathway showed that the sialic acid synthesis (but not O-GlcNac glycosylation) pathway was enhanced, which was mediated by a key enzyme, N-acetylneuraminic acid synthase. Knockout of N-acetylneuraminic acid synthase also inhibits tumor growth and extends murine survival in c-Myc-driven HCC models.

Conclusions: These findings indicate that the activation of the hexosamine biosynthetic pathway/sialic acid pathway is an important mechanism underlying the development of c-Myc-driven HCC. Inhibitors of GFPT1, along with anti- N-acetylneuraminic acid synthase may offer a promising therapeutic strategy.

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC12269569PMC
http://dx.doi.org/10.1016/j.jcmgh.2025.101523DOI Listing

Publication Analysis

Top Keywords

c-myc-driven hcc
16
murine survival
12
hexosamine biosynthetic
12
n-acetylneuraminic acid
12
acid synthase
12
hepatocellular carcinoma
8
hcc
8
rna sequencing
8
quantitative polymerase
8
polymerase chain
8

Similar Publications

Targeting c-MYC has a key role in hepatocellular carcinoma therapy.

Crit Rev Oncol Hematol

September 2025

Colorectal Surgery,Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Shan Xi Taiyuan, 030001, China. Electronic address:

Hepatocellular carcinoma (HCC) is a top cause of cancer-associated mortality worldwide, with limited effective treatment options. The oncogenic transcription factor c-MYC plays a pivotal role in HCC pathogenesis by regulating key cellular processes, including proliferation, metabolism, and apoptosis. Impaired c-MYC regulation strongly correlates with aberrant activation of multiple signaling pathways, such as PI3K/Akt/mTOR, Wnt/β-catenin, and MAPK/ERK, which collectively drive tumor progression.

View Article and Find Full Text PDF

The Role of the Hexosamine-Sialic Acid Metabolic Pathway Mediated by GFPT1/NANS in c-Myc-Driven Hepatocellular Carcinoma.

Cell Mol Gastroenterol Hepatol

August 2025

Department of Pathology, School of Basic Medical Sciences and Qilu Hospital, Shandong University, Jinan, China. Electronic address:

Background & Aims: Hepatocellular carcinoma (HCC) frequently involves metabolic reprogramming, which promotes oncogenesis and metastasis. However, the underlying molecular mechanisms remain insufficiently explored. In this study, we aim to investigate the metabolic abnormalities in c-Myc-driven HCC development and their potential therapeutic implications.

View Article and Find Full Text PDF
Article Synopsis
  • Tumor-associated neutrophils (TANs) vary in function depending on the type of cancer, with their role being more significant in metabolic dysfunction-related liver cancer than in viral-related liver cancer.* -
  • In metabolic dysfunction-associated hepatocellular carcinoma (MASH-related HCC), specific TANs (SiglecFhi) promote tumor growth and immune evasion by enhancing stemness and inhibiting the immune response.* -
  • Targeting SiglecFhi TANs can improve the effectiveness of immunotherapy, as their removal increases cancer cell recognition and correlates with poor patient outcomes due to resistance to treatment.*
View Article and Find Full Text PDF

Activated mTORC2/AKT signaling plays a role in hepatocellular carcinoma (HCC). Research has shown that TSC/mTORC1 and FOXO1 are distinct downstream effectors of AKT signaling in liver regeneration and metabolism. However, the mechanisms by which these pathways mediate mTORC2/AKT activation in HCC are not yet fully understood.

View Article and Find Full Text PDF

Mitotic arrest-deficient 2 like 1 (MAD2L1) is a component of the mitotic spindle assembly checkpoint implicated in cancer cell proliferation and tumorigenesis. The functional role of MAD2L1 in hepatocellular carcinoma (HCC) has not been adequately investigated, especially in vivo. In the current manuscript, we sought to address the function of MAD2L1 in hepatocarcinogenesis.

View Article and Find Full Text PDF