Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Gastrointest Oncol ; 12(2): 694-710, 2021 Apr.
Article in English | MEDLINE | ID: mdl-34012659

ABSTRACT

BACKGROUND: The majority of hepatocellular carcinoma (HCC) is closely associated with hepatitis B virus (HBV) infection, while the mechanism of HCC induced by HBV is debatable. Bone marrow stromal cell antigen 2 (BST-2), an N-glycoprotein, has been characterized as an oncogenic factor in several types of cancer. However, whether BST-2 plays an important role in HCC tumorigenesis remains unknown. METHODS: A total of 182 HCC tumorous and adjacent nontumor liver tissues were collected. HepG2, Huh7, L02, HepAD38, and HEK293T cell lines were adopted in this study. Tumor proliferation was detected by CCK8, transwell, wound healing, colony formation assays in vitro, and in vivo tumorigenesis was measured by mouse xenografts. NF-κB activation was determined by luciferase assay and Western blot. Protein expression was detected by Western blot, ELISA, or qPCR. Immunoprecipitation was used to confirm the interaction between BST-2 and Syk. RESULTS: Here, we observed the higher BST-2 expression in HBV-infected HCC than their paired adjacent tissues and HBV-uninfected HCC tissues, particularly more aberrant non-N-glycosylated BST-2 in HBV-infected HCC tumors. We also observed the increased ER degradation-enhancing α-mannosidase-like protein 3 (EDEM3), which is trimming of N-linked glycans by sequential removal of mannose residues, might result in more non-N-glycosylated form of BST-2. Moreover, we demonstrated that BST-2 and non-N-glycosylated BST-2 N65/92A mutant, not only enhanced the tumor characteristics of hepatoma cell lines in vitro, but also enhanced the growth of mouse xenografts in vivo. Mechanically, N65/92A mutant has stronger ability to promote HCC than BST-2 via NF-κB/ERK1/2 but not NF-κB/anti-apoptotic factors pathway. NF-κB inhibitor attenuated BST-2-mediated tumorigenesis of HCC. CONCLUSIONS: Our findings illuminate the novel function of BST-2 as an oncogene of HBV-associated HCC, and highlight the novel relationship of N-glycosylation of BST-2 in regulating HCC tumorigenesis in vitro.

2.
Virol Sin ; 36(4): 678-691, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33534085

ABSTRACT

Our recent study reported that ATP1B3 inhibits hepatitis B virus (HBV) replication via inducing NF-κB activation. However, ATP1B3 mutants which were defective in NF-κB activation still maintained the moderate degree of suppression on HBV replication, suggesting that another uncharacterized mechanism is also responsible for ATP1B3-mediated HBV suppression. Here, we demonstrated that ATP1B3 reduced the expression of HBV envelope proteins LHBs, MHBs and SHBs, but had no effect on intracellular HBV DNA, RNA levels as well as HBV promoter activities. Further investigation showed that proteasome inhibitor MG132 rescued ATP1B3-mediated envelope proteins degradation, demonstrating that proteasome-dependent pathway is involved in ATP1B3-induced degradation of envelope proteins. Co-IP showed that ATP1B3 interacts with LHBs and MHBs and induces LHBs and MHBs polyubiquitination. Immunofluorescence co-localization analysis confirmed LHBs and MHBs colocalized with ATP1B3 together. Our work provides important information for targeting ATP1B3 as a potential therapeutic molecule for HBV infection.


Subject(s)
Hepatitis B virus , Hepatitis B , Gene Products, env , Hepatitis B Surface Antigens , Humans , Sodium-Potassium-Exchanging ATPase , Virus Replication
3.
Front Microbiol ; 11: 697, 2020.
Article in English | MEDLINE | ID: mdl-32431673

ABSTRACT

Serine incorporator 3 (SERINC3) and SERINC5 were recently identified as host intrinsic factors against human immunodeficiency virus (HIV)-1 and counteracted by HIV-1 Nef. However, whether they inhibit hepatitis B virus (HBV), which is a severe health problem worldwide, is unknown. Here, we demonstrate that SERINC5 potently inhibited HBV virion secretion in the supernatant without affecting intracellular core particle-associated DNA and the total RNA, but SERINC3 and SERINC1 did not. Further investigation discovered that SERINC5 increased the non-glycosylation of LHB, MHB, and SHB proteins of HBV and slightly decreased HBs proteins levels, which led to the decreased HBV secretion. Importantly, SERINC5 co-localized with LHB proteins in the Golgi apparatus, which is important for glycan processing and transport. In addition, we determined the functional domain in SERINC5 required for HBV inhibition, which was completely different from that required for HIV-1 restriction, whereas phosphorylation and glycosylation sites in SERINC5 were dispensable for HBV restriction. Taken together, our results demonstrate that SERINC5 suppresses HBV virion secretion through interfering with the glycosylation of HBV proteins, suggesting that SERINC5 might possess broad-spectrum antiviral activity.

4.
J Med Virol ; 92(2): 201-209, 2020 02.
Article in English | MEDLINE | ID: mdl-31556466

ABSTRACT

Increasing evidence indicates ATP1B3, one of the regulatory subunits of Na+ /K+ -ATPase, is involved in numerous viral propagations, such as HIV and EV71. However, the function and mechanism of ATP1B3 on hepatitis B virus (HBV) propagation is unknown. Here, we demonstrated that ATP1B3 overexpression reduced the quantity of hepatitis B surface antigen (HBsAg) and hepatitis B e antigen (HBeAg) in supernatants of HBV expression plasmids cotransfected HepG2 cells. Correspondingly, small interfering RNA and short hairpin RNA mediated ATP1B3 silencing promoted HBsAg and HBeAg expression in the supernatants of HBV expression plasmids transfected HepG2 cells. Mechanically, we reported that ATP1B3 expression could activate nuclear factor-κB (NF-κB) pathway by inducing the expression, phosphorylation, and nuclear import of P65 for the first time. And NF-κB inhibitor (Bay11) impaired the restraint of ATP1B3 on HBV replication. This counteraction effect of Bay11 proved that ATP1B3-induced NF-κB activation was crucial for HBV restriction. Accordingly, we observed that anti-HBV factors interferon-α (IFN-α) and interleukin-6 (IL-6) production were increased in HepG2 cells after the NF-κB activation. It suggested that ATP1B3 suppressed HBsAg and HBeAg by NF-κB/IFN-α and NF-κB/IL-6 axis. Further experiments proved that ATP1B3 overexpression induced anti-HBV factor BST-2 expression by NF-κB/IFN-α axis in HepG2 cells but not HEK293T cells, and ATP1B3 silencing downregulated BST-2 messenger RNA level in HepG2 cells. As an HBV restriction factor, BST-2 cooperated with ATP1B3 to antagonize HBsAg but not HBeAg in HepG2 cells. Our work identified ATP1B3 as a novel candidate of HBV restrictor with unrevealed mechanism and we highlighted it might serve as a potential therapeutic molecule for HBV infection.


Subject(s)
Antigens, CD/metabolism , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatitis B virus/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Adaptor Proteins, Signal Transducing , Antigens, CD/genetics , Cell Survival , Cytokines/genetics , Cytokines/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Viral/physiology , HEK293 Cells , Hep G2 Cells , Hepatitis B Surface Antigens/genetics , Hepatitis B e Antigens/genetics , Humans , Interferon-alpha/genetics , Interferon-alpha/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , RNA-Binding Proteins , Sodium-Potassium-Exchanging ATPase/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...