File Download
Supplementary

postgraduate thesis: C-terminal truncated HBx initiates hepatocarcinogenesis by down-regulating TXNIP and reprogramming glucose metabolism

TitleC-terminal truncated HBx initiates hepatocarcinogenesis by down-regulating TXNIP and reprogramming glucose metabolism
Authors
Advisors
Advisor(s):Guan, XLee, VHF
Issue Date2021
PublisherThe University of Hong Kong (Pokfulam, Hong Kong)
Citation
Zhang, Y. [張渝]. (2021). C-terminal truncated HBx initiates hepatocarcinogenesis by down-regulating TXNIP and reprogramming glucose metabolism. (Thesis). University of Hong Kong, Pokfulam, Hong Kong SAR.
AbstractChronic hepatitis B virus (HBV) infection is the leading causative factor for hepatocellular carcinoma (HCC) , accounting for more than 60% HCC cases in east Asian countries including China. HBV integration into the host genome has been proven strongly associated with the initiation and progression of HCC. However, a substantial amount of virus-induced mechanisms that cause the malignant transformation of HCC cells remains largely unknown, especially for the HBV-induced glucose metabolic reprogramming in HCC. In the present study, we firstly analyzed the distribution of integration breakpoints in the virus genome from the whole genome sequencing of HBV-related HCC in public databases. We found that HBV integration breakpoints clustered around the 3′-end of HBV X gene in HCC samples, thus we hypothesized that the integration associated C-terminal truncated HBx (Ct-HBx) protein is oncogenic in tumorigenesis. In vitro and in vivo functional assays were applied to study the role of Ct-HBx in HCC cells. Transcriptome sequencing was performed to characterize the molecular basis of the Ct-HBx induced HCC progression. Ct-HBx is more frequently expressed in HCC samples than the full-length HBx. The oncogenic properties of Ct-HBx were validated in both vitro and vivo functional assays. RNA-sequencing suggested that Ct-HBx mediated the glucose metabolic alteration from oxidative phosphorylation to glycolysis, which is the hallmark of cancer progression. RNA-sequencing also identified TXNIP as the potential target of Ct-HBx, which may mediate metabolic reprogramming. Bioinformatic data from HCC samples in both TCGA database and our own cohort exhibited that TXNIP is significantly downregulated in HCC patients with Ct-HBx expression, compared to those with full-length HBx expression and HBV-negative samples. Lower expression of TXNIP is associated with poor prognosis. We also showed that Ct-HBx transcriptionally downregulates TXNIP through transactivation. Additionally, re-introduction of TXNIP inhibited the metabolic alterations induced by Ct-HBx, thus resulting in tumor growth arrest in HCC. Our data demonstrated that the generation of C-terminal truncated HBx upon HBV integration has an oncogenic role in HCC development. Ct-HBx promotes glycolysis metabolism through downregulating the expression of TXNIP. Taken together, our finding proposes a promising diagnostic and therapeutic target for HBV-induced HCC.
DegreeDoctor of Philosophy
SubjectHepatitis B virus
Liver - Cancer
Dept/ProgramClinical Oncology
Persistent Identifierhttp://hdl.handle.net/10722/306990

 

DC FieldValueLanguage
dc.contributor.advisorGuan, X-
dc.contributor.advisorLee, VHF-
dc.contributor.authorZhang, Yu-
dc.contributor.author張渝-
dc.date.accessioned2021-11-03T04:36:39Z-
dc.date.available2021-11-03T04:36:39Z-
dc.date.issued2021-
dc.identifier.citationZhang, Y. [張渝]. (2021). C-terminal truncated HBx initiates hepatocarcinogenesis by down-regulating TXNIP and reprogramming glucose metabolism. (Thesis). University of Hong Kong, Pokfulam, Hong Kong SAR.-
dc.identifier.urihttp://hdl.handle.net/10722/306990-
dc.description.abstractChronic hepatitis B virus (HBV) infection is the leading causative factor for hepatocellular carcinoma (HCC) , accounting for more than 60% HCC cases in east Asian countries including China. HBV integration into the host genome has been proven strongly associated with the initiation and progression of HCC. However, a substantial amount of virus-induced mechanisms that cause the malignant transformation of HCC cells remains largely unknown, especially for the HBV-induced glucose metabolic reprogramming in HCC. In the present study, we firstly analyzed the distribution of integration breakpoints in the virus genome from the whole genome sequencing of HBV-related HCC in public databases. We found that HBV integration breakpoints clustered around the 3′-end of HBV X gene in HCC samples, thus we hypothesized that the integration associated C-terminal truncated HBx (Ct-HBx) protein is oncogenic in tumorigenesis. In vitro and in vivo functional assays were applied to study the role of Ct-HBx in HCC cells. Transcriptome sequencing was performed to characterize the molecular basis of the Ct-HBx induced HCC progression. Ct-HBx is more frequently expressed in HCC samples than the full-length HBx. The oncogenic properties of Ct-HBx were validated in both vitro and vivo functional assays. RNA-sequencing suggested that Ct-HBx mediated the glucose metabolic alteration from oxidative phosphorylation to glycolysis, which is the hallmark of cancer progression. RNA-sequencing also identified TXNIP as the potential target of Ct-HBx, which may mediate metabolic reprogramming. Bioinformatic data from HCC samples in both TCGA database and our own cohort exhibited that TXNIP is significantly downregulated in HCC patients with Ct-HBx expression, compared to those with full-length HBx expression and HBV-negative samples. Lower expression of TXNIP is associated with poor prognosis. We also showed that Ct-HBx transcriptionally downregulates TXNIP through transactivation. Additionally, re-introduction of TXNIP inhibited the metabolic alterations induced by Ct-HBx, thus resulting in tumor growth arrest in HCC. Our data demonstrated that the generation of C-terminal truncated HBx upon HBV integration has an oncogenic role in HCC development. Ct-HBx promotes glycolysis metabolism through downregulating the expression of TXNIP. Taken together, our finding proposes a promising diagnostic and therapeutic target for HBV-induced HCC.-
dc.languageeng-
dc.publisherThe University of Hong Kong (Pokfulam, Hong Kong)-
dc.relation.ispartofHKU Theses Online (HKUTO)-
dc.rightsThe author retains all proprietary rights, (such as patent rights) and the right to use in future works.-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subject.lcshHepatitis B virus-
dc.subject.lcshLiver - Cancer-
dc.titleC-terminal truncated HBx initiates hepatocarcinogenesis by down-regulating TXNIP and reprogramming glucose metabolism-
dc.typePG_Thesis-
dc.description.thesisnameDoctor of Philosophy-
dc.description.thesislevelDoctoral-
dc.description.thesisdisciplineClinical Oncology-
dc.description.naturepublished_or_final_version-
dc.date.hkucongregation2021-
dc.identifier.mmsid991044437602303414-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats