Abstract

Chronic hepatitis B virus (HBV) infection is strongly associated with the initiation and development of hepatocellular carcinoma (HCC). However, the genetic alterations and pathogenesis mechanisms remain significantly unexplored, especially for HBV-induced metabolic reprogramming. Analysis of integration breakpoints in HBV-positive HCC samples revealed the preferential clustering pattern within the 3′-end of X gene in the HBV genome, leading to the production of C-terminal truncated X protein (Ct-HBx). In this study, we not only characterized the oncogenic role of two Ct-HBx (HBx-120 and HBx-134) via in vitro and in vivo functional assays but also deciphered their underlying molecular mechanisms. Gene expression profiling by transcriptome sequencing identified potential targets of Ct-HBx and novel malignant hallmarks such as glycolysis, cell cycle, and m-TORC1 signaling in Ct-HBx-expressing cells. TXNIP, a well-established regulator of glucose metabolism, was shown to be downregulated by Ct-HBx and play a pivotal role in Ct-HBx-mediated HCC progression. Suppression of TXNIP is frequently observed in HCC patients with Ct-HBx expression and significantly (P = 0.015) correlated to a poorer prognosis. Re-introduction of TXNIP attenuated the metabolic reprogramming induced by the Ct-HBx and inhibited the tumor growth in the mice model. Further study suggested that Ct-HBx could downregulate TXNIP via a transcriptional repressor nuclear factor of activated T cells 2 (NFACT2). Collectively, our findings indicate that TXNIP plays a critical role in Ct-HBx-mediated hepatocarcinogenesis, serving as a novel therapeutic strategy in HCC treatment.

Highlights

  • These authors contributed : Yu Zhang, Qian Yan

  • In order to examine the precise distribution of hepatitis B virus (HBV) integration breakpoints, we analyzed the whole-genome sequencing (WGS) data for 29 hepatocellular carcinoma (HCC) samples from ERP001196 in the European Genomephenome Archive (EGA) [6]

  • Single-cell genome sequencing has confirmed that in monoclonal HCC models, HBV integration pattern remains extremely unchanged during hepatocarcinogenesis

Read more

Summary

1234567890();,: 1234567890();,: Introduction

The emerging advances suggest that HCC progression involves multiple HBV-induced alterations including liver fibrosis/cirrhosis [4, 5], genomic integration of viral DNA [6, 7], and the oncogenic potential of HBVencoding proteins, such as X protein (HBx) [8]. Based on the transcriptomics screening and profiling, thioredoxin interacting protein (TXNIP) has been identified and characterized as the downstream target of Ct-HBx, acting as a negative regulator of aerobic glycolysis during HCC progression. HCC is characterized as one of the most hypoxic solid tumors with the average oxygen content of 0.8%, little is known about the metabolic-regulating function of TXNIP in HBV-induced HCC. In the present study, integrated bioinformatics analysis of whole-genome sequence and both in vivo and in vitro functional assay confirmed the oncogenic function of Ct-HBx in HBV-induced HCC. We demonstrated that Ct-HBx transactivates NFATC2, facilitating the transcriptional repression of TXNIP

Results
Discussion
Materials and methods
Compliance with ethical standards
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call