Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.441
Filter
1.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38928309

ABSTRACT

Hepatitis B virus (HBV) infects approximately 300 million people worldwide, causing chronic infections. The HBV X protein (HBx) is crucial for viral replication and induces reactive oxygen species (ROS), leading to cellular damage. This study explores the relationship between HBx-induced ROS, p53 activation, and HBV replication. Using HepG2 and Hep3B cell lines that express the HBV receptor NTCP, we compared ROS generation and HBV replication relative to p53 status. Results indicated that HBV infection significantly increased ROS levels in p53-positive HepG2-NTCP cells compared to p53-deficient Hep3B-NTCP cells. Knockdown of p53 reduced ROS levels and enhanced HBV replication in HepG2-NTCP cells, whereas p53 overexpression increased ROS and inhibited HBV replication in Hep3B-NTCP cells. The ROS scavenger N-acetyl-L-cysteine (NAC) reversed these effects. The study also found that ROS-induced degradation of the HBx is mediated by the E3 ligase Siah-1, which is activated by p53. Mutations in p53 or inhibition of its transcriptional activity prevented ROS-mediated HBx degradation and HBV inhibition. These findings reveal a p53-dependent negative feedback loop where HBx-induced ROS increases p53 levels, leading to Siah-1-mediated HBx degradation and HBV replication inhibition. This study offers insights into the molecular mechanisms of HBV replication and identifies potential therapeutic targets involving ROS and p53 pathways.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B virus , Liver Neoplasms , Reactive Oxygen Species , Trans-Activators , Tumor Suppressor Protein p53 , Ubiquitin-Protein Ligases , Viral Regulatory and Accessory Proteins , Virus Replication , Humans , Tumor Suppressor Protein p53/metabolism , Hepatitis B virus/physiology , Reactive Oxygen Species/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Hep G2 Cells , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Liver Neoplasms/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Line, Tumor
2.
Biomolecules ; 14(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38927063

ABSTRACT

The Ebola virus (EBOV) is a lethal pathogen causing hemorrhagic fever syndrome which remains a global health challenge. In the EBOV, two multifunctional proteins, VP35 and VP40, have significant roles in replication, virion assembly, and budding from the cell and have been identified as druggable targets. In this study, we employed in silico methods comprising molecular docking, molecular dynamic simulations, and pharmacological properties to identify prospective drugs for inhibiting VP35 and VP40 proteins from the myxobacterial bioactive natural product repertoire. Cystobactamid 934-2, Cystobactamid 919-1, and Cittilin A bound firmly to VP35. Meanwhile, 2-Hydroxysorangiadenosine, Enhypyrazinone B, and Sorangiadenosine showed strong binding to the matrix protein VP40. Molecular dynamic simulations revealed that, among these compounds, Cystobactamid 919-1 and 2-Hydroxysorangiadenosine had stable interactions with their respective targets. Similarly, molecular mechanics Poisson-Boltzmann surface area (MMPBSA) calculations indicated close-fitting receptor binding with VP35 or VP40. These two compounds also exhibited good pharmacological properties. In conclusion, we identified Cystobactamid 919-1 and 2-Hydroxysorangiadenosine as potential ligands for EBOV that target VP35 and VP40 proteins. These findings signify an essential step in vitro and in vivo to validate their potential for EBOV inhibition.


Subject(s)
Antiviral Agents , Biological Products , Ebolavirus , Molecular Docking Simulation , Molecular Dynamics Simulation , Ebolavirus/drug effects , Biological Products/pharmacology , Biological Products/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Myxococcales/chemistry , Humans , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/chemistry , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/chemistry , Nucleocapsid Proteins
3.
J Cell Mol Med ; 28(11): e18484, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38842124

ABSTRACT

As an important protein encoded by hepatitis B virus (HBV), HBV X protein (HBx) plays an important role in the development of hepatocellular carcinoma (HCC). It has been shown that seven in absentia homologue 1 (SIAH1) could regulates the degradation of HBx through the ubiquitin-proteasome pathway. However, as a member of SIAH family, the regulatory effects of SIAH2 on HBx remain unclear. In this study, we first confirmed that SIAH2 could reduce the protein levels of HBx depending on its E3 ligase activity. Moreover, SIAH2 interacted with HBx and induced its K48-linked polyubiquitination and proteasomal degradation. Furthermore, we provided evidence that SIAH2 inhibits HBx-associated HCC cells proliferation by regulating HBx. In conclusion, our study identified a novel role for SIAH2 in promoting HBx degradation and SIAH2 exerts an inhibitory effect in the proliferation of HBx-associated HCC through inducing the degradation of HBx. Our study provides a new idea for the targeted degradation of HBx and may have great huge significance into providing novel evidence for the targeted therapy of HBV-infected HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Hepatitis B virus , Liver Neoplasms , Nuclear Proteins , Proteolysis , Trans-Activators , Ubiquitin-Protein Ligases , Ubiquitination , Viral Regulatory and Accessory Proteins , Humans , Viral Regulatory and Accessory Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Trans-Activators/metabolism , Trans-Activators/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , Liver Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Hepatitis B virus/metabolism , Hepatitis B virus/pathogenicity , Cell Line, Tumor , Signal Transduction , Hep G2 Cells
4.
PLoS One ; 19(6): e0305350, 2024.
Article in English | MEDLINE | ID: mdl-38861553

ABSTRACT

All-trans retinoic acid (ATRA), recognized as the principal and most biologically potent metabolite of vitamin A, has been identified for its inhibitory effects on hepatitis B virus (HBV) replication. Nevertheless, the underlying mechanism remains elusive. The present study reveals that ATRA induces E6-associated protein (E6AP)-mediated proteasomal degradation of HBx to suppress HBV replication in human hepatoma cells in a p53-dependent pathway. For this effect, ATRA induced promoter hypomethylation of E6AP in the presence of HBx, which resulted in the upregulation of E6AP levels in HepG2 but not in Hep3B cells, emphasizing the p53-dependent nature of this effect. As a consequence, ATRA augmented the interaction between E6AP and HBx, resulting in substantial ubiquitination of HBx and consequent reduction in HBx protein levels in both the HBx overexpression system and the in vitro HBV replication model. Additionally, the knockdown of E6AP under ATRA treatment reduced the interaction between HBx and E6AP and decreased the ubiquitin-dependent proteasomal degradation of HBx, which prompted a recovery of HBV replication in the presence of ATRA, as confirmed by increased levels of intracellular HBV proteins and secreted HBV levels. This study not only contributes to the understanding of the complex interactions between ATRA, p53, E6AP, and HBx but also provides an academic basis for the clinical employment of ATRA in the treatment of HBV infection.


Subject(s)
Hepatitis B virus , Proteasome Endopeptidase Complex , Trans-Activators , Tretinoin , Tumor Suppressor Protein p53 , Ubiquitin-Protein Ligases , Viral Regulatory and Accessory Proteins , Virus Replication , Humans , Viral Regulatory and Accessory Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Proteasome Endopeptidase Complex/metabolism , Virus Replication/drug effects , Hepatitis B virus/physiology , Hepatitis B virus/drug effects , Tretinoin/pharmacology , Tretinoin/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Hep G2 Cells , Down-Regulation/drug effects , Ubiquitination/drug effects , Proteolysis/drug effects , Promoter Regions, Genetic , DNA Methylation/drug effects , Cell Line, Tumor
5.
J Zhejiang Univ Sci B ; 25(6): 499-512, 2024 Jun 01.
Article in English, Chinese | MEDLINE | ID: mdl-38910495

ABSTRACT

Artificial vascular graft (AVG) fistula is widely used for hemodialysis treatment in patients with renal failure. However, it has poor elasticity and compliance, leading to stenosis and thrombosis. The ideal artificial blood vessel for dialysis should replicate the structure and components of a real artery, which is primarily maintained by collagen in the extracellular matrix (ECM) of arterial cells. Studies have revealed that in hepatitis B virus (HBV)-induced liver fibrosis, hepatic stellate cells (HSCs) become hyperactive and produce excessive ECM fibers. Furthermore, mechanical stimulation can encourage ECM secretion and remodeling of a fiber structure. Based on the above factors, we transfected HSCs with the hepatitis B viral X (HBX) gene for simulating the process of HBV infection. Subsequently, these HBX-HSCs were implanted into a polycaprolactone-polyurethane (PCL-PU) bilayer scaffold in which the inner layer is dense and the outer layer consists of pores, which was mechanically stimulated to promote the secretion of collagen nanofiber from the HBX-HSCs and to facilitate crosslinking with the scaffold. We obtained an ECM-PCL-PU composite bionic blood vessel that could act as access for dialysis after decellularization. Then, the vessel scaffold was implanted into a rabbit's neck arteriovenous fistula model. It exhibited strong tensile strength and smooth blood flow and formed autologous blood vessels in the rabbit's body. Our study demonstrates the use of human cells to create biomimetic dialysis blood vessels, providing a novel approach for creating clinical vascular access for dialysis.


Subject(s)
Hepatic Stellate Cells , Polyesters , Renal Dialysis , Rabbits , Animals , Polyesters/chemistry , Viral Regulatory and Accessory Proteins , Tissue Scaffolds , Transfection , Bionics , Polyurethanes , Blood Vessel Prosthesis , Extracellular Matrix/metabolism , Humans , Hepatitis B virus/genetics , Collagen , Tissue Engineering/methods , Trans-Activators
6.
Virology ; 595: 110065, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38569227

ABSTRACT

Nucleot(s)ide analogues, the current antiviral treatments against chronic hepatitis B (CHB) infection, are non-curative due to their inability to eliminate covalently closed circular DNA (cccDNA) from the infected hepatocytes. Preclinical studies have shown that coumarin derivatives can effectively reduce the HBV DNA replication. We evaluated the antiviral efficacy of thirty new coumarin derivatives in cell culture models for studying HBV. Furanocoumarins Fc-20 and Fc-31 suppressed the levels of pre-genomic RNA as well as cccDNA, and reduced the secretion of virions, HBsAg and HBeAg. The antiviral efficacies of Fc-20 and Fc31 improved further when used in combination with the hepatitis B antiviral drug Entecavir. There was a marked reduction in the intracellular HBx level in the presence of these furanocoumarins due to proteasomal degradation resulting in the down-regulation of HBx-dependent viral genes. Importantly, both Fc-20 and Fc-31 were non-cytotoxic to cells even at high concentrations. Further, our molecular docking studies confirmed a moderate to high affinity interaction between furanocoumarins and viral HBx via residues Ala3, Arg26 and Lys140. These data suggest that furanocoumarins could be developed as a new therapeutic for CHB infection.


Subject(s)
Antiviral Agents , DNA, Circular , Furocoumarins , Hepatitis B virus , Proteasome Endopeptidase Complex , Trans-Activators , Viral Regulatory and Accessory Proteins , Virus Replication , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B virus/metabolism , Virus Replication/drug effects , Humans , Trans-Activators/metabolism , Trans-Activators/genetics , DNA, Circular/metabolism , DNA, Circular/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Furocoumarins/pharmacology , Antiviral Agents/pharmacology , Proteasome Endopeptidase Complex/metabolism , DNA, Viral/metabolism , DNA, Viral/genetics , Down-Regulation/drug effects , Transcription, Genetic/drug effects , Proteolysis/drug effects , Gene Expression Regulation, Viral/drug effects , Hep G2 Cells
7.
Cell Signal ; 119: 111183, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636768

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality worldwide, with Hepatitis B virus (HBV) infection being the leading cause. This study aims to investigate the role of HBV in HCC pathogenesis involving glucose metabolism. Long non-coding RNA (lncRNA) OIP5-AS1 was significantly downregulated in HBV-positive HCC patients, and its low expression indicated a poor prognosis. This lncRNA was primarily localized in the cytoplasm, acting as a tumor suppressor. HBV protein X (HBx) repressed OIP5-AS1 expression by inhibiting a ligand-activated transcriptional factor peroxisome proliferator-activated receptor α (PPARα). Furthermore, mechanistic studies revealed that OIP5-AS1 inhibited tumor growth by suppressing Hexokinase domain component 1 (HKDC1)-mediated glycolysis. The expression of HKDC1 could be enhanced by transcriptional factor sterol regulatory element-binding protein 1 (SREBP1). OIP5-AS1 facilitated the ubiquitination and degradation of SREBP1 to suppress HKDC1 transcription, which inhibited glycolysis. The results suggest that lncRNA OIP5-AS1 plays an anti-oncogenic role in HBV-positive HCC via the HBx/OIP5-AS1/HKDC1 axis, providing a promising diagnostic marker and therapeutic target for HBV-positive HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Gene Expression Regulation, Neoplastic , Glycolysis , Hexokinase , Liver Neoplasms , RNA, Long Noncoding , Trans-Activators , Viral Regulatory and Accessory Proteins , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/virology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Glycolysis/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Hexokinase/metabolism , Hexokinase/genetics , Animals , Hepatitis B virus , Male , Cell Line, Tumor , Down-Regulation , Mice , Mice, Nude , Female , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Mice, Inbred BALB C , PPAR alpha/metabolism , PPAR alpha/genetics
8.
J Virol ; 98(5): e0157323, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38572974

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and characterized by dysregulated immune response. Studies have shown that the SARS-CoV-2 accessory protein ORF7b induces host cell apoptosis through the tumor necrosis factor alpha (TNF-α) pathway and blocks the production of interferon beta (IFN-ß). The underlying mechanism remains to be investigated. In this study, we found that ORF7b facilitated viral infection and production, and inhibited the RIG-I-like receptor (RLR) signaling pathway through selectively interacting with mitochondrial antiviral-signaling protein (MAVS). MAVS439-466 region and MAVS Lys461 were essential for the physical association between MAVS and ORF7b, and the inhibition of the RLR signaling pathway by ORF7b. MAVSK461/K63 ubiquitination was essential for the RLR signaling regulated by the MAVS-ORF7b complex. ORF7b interfered with the recruitment of tumor necrosis factor receptor-related factor 6 (TRAF6) and the activation of the RLR signaling pathway by MAVS. Furthermore, interfering peptides targeting the ORF7b complex reversed the ORF7b-suppressed MAVS-RLR signaling pathway. The most potent interfering peptide V disrupts the formation of ORF7b tetramers, reverses the levels of the ORF7b-inhibited physical association between MAVS and TRAF6, leading to the suppression of viral growth and infection. Overall, this study provides a mechanism for the suppression of innate immunity by SARS-CoV-2 infection and the mechanism-based approach via interfering peptides to potentially prevent SARS-CoV-2 infection.IMPORTANCEThe pandemic coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and continues to be a threat to public health. It is imperative to understand the biology of SARS-CoV-2 infection and find approaches to prevent SARS-CoV-2 infection and ameliorate COVID-19. Multiple SARS-CoV-2 proteins are known to function on the innate immune response, but the underlying mechanism remains unknown. This study shows that ORF7b inhibits the RIG-I-like receptor (RLR) signaling pathway through the physical association between ORF7b and mitochondrial antiviral-signaling protein (MAVS), impairing the K63-linked MAVS polyubiquitination and its recruitment of tumor necrosis factor receptor-related factor 6 (TRAF6) to MAVS. The most potent interfering peptide V targeting the ORF7b-MAVS complex may reverse the suppression of the MAVS-mediated RLR signaling pathway by ORF7b and prevent viral infection and production. This study may provide new insights into the pathogenic mechanism of SARS-CoV-2 and a strategy to develop new drugs to prevent SARS-CoV-2 infection.


Subject(s)
Adaptor Proteins, Signal Transducing , COVID-19 , SARS-CoV-2 , Signal Transduction , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , COVID-19/virology , COVID-19/immunology , COVID-19/metabolism , DEAD Box Protein 58/metabolism , HEK293 Cells , Immunity, Innate , Interferon-beta/metabolism , Receptors, Immunologic/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics
9.
J Virol ; 98(5): e0042424, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38629837

ABSTRACT

Chronic hepatitis B virus (HBV) infections are strongly associated with liver cirrhosis, inflammation, and hepatocellular carcinoma. In this context, the viral HBx protein is considered as a major factor influencing HBV-associated pathogenesis through deregulation of multiple cellular signaling pathways and is therefore a potential target for prognostic and therapeutic applications. However, HBV-associated pathogenesis differs significantly between genotypes, with the relevant factors and in particular the contribution of the genetic diversity of HBx being largely unknown. To address this question, we studied the specific genotype-dependent impact of HBx on cellular signaling pathways, focusing in particular on morphological and functional parameters of mitochondria. To exclusively investigate the impact of HBx of different genotypes on integrity and function of mitochondria in the absence of additional viral factors, we overexpressed HBx in Huh7 or HepG2 cells. Key signaling pathways were profiled by kinome analysis and correlated with expression levels of mitochondrial and pathogenic markers. Conclusively, HBx of genotypes A and G caused strong disruption of mitochondrial morphology alongside an induction of PTEN-induced putative kinase 1/Parkin-mediated mitophagy. These effects were only moderately dysregulated by genotypes B and E, whereas genotypes C and D exhibit an intermediate effect in this regard. Accordingly, changes in mitochondrial membrane potential and elevated reactive oxygen species production were associated with the HBx-mediated dysfunction among different genotypes. Also, genotype-related differences in mitophagy induction were identified and indicated that HBx-mediated changes in the mitochondria morphology and function strongly depend on the genotype. This indicates a relevant role of HBx in the process of genotype-dependent liver pathogenesis of HBV infections and reveals underlying mechanisms.IMPORTANCEThe hepatitis B virus is the main cause of chronic liver disease worldwide and differs in terms of pathogenesis and clinical outcome among the different genotypes. Furthermore, the viral HBx protein is a known factor in the progression of liver injury by inducing aberrant mitochondrial structures and functions. Consequently, the selective removal of dysfunctional mitochondria is essential to maintain overall cellular homeostasis and cell survival. Consistent with the intergenotypic difference of HBV, our data reveal significant differences regarding the impact of HBx of different genotypes on mitochondrial dynamic and function and thereby on radical oxygen stress levels within the cell. We subsequently observed that the induction of mitophagy differs significantly across the heterogenetic HBx proteins. Therefore, this study provides evidence that HBx-mediated changes in the mitochondria dynamics and functionality strongly depend on the genotype of HBx. This highlights an important contribution of HBx in the process of genotype-dependent liver pathogenesis.


Subject(s)
Hepatitis B virus , Mitochondrial Dynamics , Signal Transduction , Trans-Activators , Viral Regulatory and Accessory Proteins , Humans , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Genotype , Hep G2 Cells , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/pathology , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitophagy , Reactive Oxygen Species/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Viral Regulatory and Accessory Proteins/metabolism
10.
Biochem Biophys Res Commun ; 712-713: 149945, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38640732

ABSTRACT

ORF3b is one of the SARS-CoV-2 accessory proteins. Previous experimental study suggested that ORF3b prevents IRF3 translocating to nucleus. However, the biophysical mechanism of ORF3b-IRF3 interaction is elusive. Here, we explored the conformation ensemble of ORF3b using all-atom replica exchange molecular dynamics simulation. Disordered ORF3b has mixed α-helix, ß-turn and loop conformers. The potential ORF3b-IRF3 binding modes were searched by docking representative ORF3b conformers with IRF3, and 50 ORF3b-IRF3 complex poses were screened using molecular dynamics simulations ranging from 500 to 1000 ns. We found that ORF3b binds IRF3 predominantly on its CBP binding and phosphorylated pLxIS motifs, with CBP binding site has the highest binding affinity. The ORF3b-IRF3 binding residues are highly conserved in SARS-CoV-2. Our results provided biophysics insights into ORF3b-IRF3 interaction and explained its interferon antagonism mechanism.


Subject(s)
Interferon Regulatory Factor-3 , Molecular Dynamics Simulation , Protein Binding , SARS-CoV-2 , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/chemistry , SARS-CoV-2/metabolism , SARS-CoV-2/chemistry , Humans , Binding Sites , COVID-19/virology , COVID-19/metabolism , Molecular Docking Simulation , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/chemistry , Protein Conformation
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167132, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565386

ABSTRACT

The Epstein-Barr virus (EBV) is implicated in several cancers, including EBV-associated gastric cancer (EBVaGC). This study focuses on EBV-encoded BALF1 (BamH1 A fragment leftward reading frame 1), a key apoptosis regulator in EBV-related cancers, whose specific impact on EBVaGC was previously unknown. Our findings indicate that BALF1 overexpression in gastric cancer cells significantly enhances their proliferation, migration, and resistance to chemotherapy-induced apoptosis, confirming BALF1's oncogenic potential. A novel discovery is that BALF1 undergoes degradation via the ubiquitin-proteasome pathway. Through analysis of 69 deubiquitinating enzymes (DUBs), ovarian tumor protease (OTU) domain-containing protein 1 (OTUD1) emerged as a vital regulator for maintaining BALF1 protein stability. Furthermore, BALF1 was found to play a role in regulating the stability of the B-cell lymphoma-2 (Bcl-2) protein, increasing its levels through deubiquitination. This mechanism reveals BALF1's multifaceted oncogenic role in gastric cancer, as it contributes both directly and indirectly to cancer progression, particularly by stabilizing Bcl-2, known for its anti-apoptotic characteristics. These insights significantly deepen our understanding of EBV's involvement in the pathogenesis of gastric cancer. The elucidation of OTUD1's role in BALF1 regulation and its influence on Bcl-2 stabilization provide new avenues for therapeutic intervention in EBVaGC, bridging the gap between viral oncogenesis and cellular protein regulation and offering a more holistic view of gastric cancer development under the influence of EBV.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2 , Stomach Neoplasms , Ubiquitination , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Cell Line, Tumor , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Cell Proliferation , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/genetics , Protein Stability , Cell Movement , Animals , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Viral Regulatory and Accessory Proteins
12.
Nucleic Acids Res ; 52(11): 6441-6458, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38499483

ABSTRACT

Coronaviruses modify their single-stranded RNA genome with a methylated cap during replication to mimic the eukaryotic mRNAs. The capping process is initiated by several nonstructural proteins (nsp) encoded in the viral genome. The methylation is performed by two methyltransferases, nsp14 and nsp16, while nsp10 acts as a co-factor to both. Additionally, nsp14 carries an exonuclease domain which operates in the proofreading system during RNA replication of the viral genome. Both nsp14 and nsp16 were reported to independently bind nsp10, but the available structural information suggests that the concomitant interaction between these three proteins would be impossible due to steric clashes. Here, we show that nsp14, nsp10, and nsp16 can form a heterotrimer complex upon significant allosteric change. This interaction is expected to encourage the formation of mature capped viral mRNA, modulating nsp14's exonuclease activity, and protecting the viral RNA. Our findings show that nsp14 is amenable to allosteric regulation and may serve as a novel target for therapeutic approaches.


Subject(s)
Methyltransferases , RNA, Viral , SARS-CoV-2 , Viral Nonstructural Proteins , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/chemistry , Methyltransferases/metabolism , Methyltransferases/genetics , Methyltransferases/chemistry , Methylation , RNA, Viral/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Exoribonucleases/metabolism , Exoribonucleases/genetics , Humans , Protein Binding , RNA Caps/metabolism , RNA Caps/genetics , Allosteric Regulation , COVID-19/virology , COVID-19/genetics , Protein Multimerization , Virus Replication/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA, Messenger/chemistry , Viral Regulatory and Accessory Proteins
13.
J Am Soc Mass Spectrom ; 35(5): 912-921, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38535992

ABSTRACT

Structure-based drug design, which relies on precise understanding of the target protein and its interaction with the drug candidate, is dramatically expedited by advances in computational methods for candidate prediction. Yet, the accuracy needs to be improved with more structural data from high throughput experiments, which are challenging to generate, especially for dynamic and weak associations. Herein, we applied native mass spectrometry (native MS) to rapidly characterize ligand binding of an allosteric heterodimeric complex of SARS-CoV-2 nonstructural proteins (nsp) nsp10 and nsp16 (nsp10/16), a complex essential for virus survival in the host and thus a desirable drug target. Native MS showed that the dimer is in equilibrium with monomeric states in solution. Consistent with the literature, well characterized small cosubstrate, RNA substrate, and product bind with high specificity and affinity to the dimer but not the free monomers. Unsuccessfully designed ligands bind indiscriminately to all forms. Using neutral gas collision, the nsp16 monomer with bound cosubstrate can be released from the holo dimer complex, confirming the binding to nsp16 as revealed by the crystal structure. However, we observed an unusual migration of the endogenous zinc ions bound to nsp10 to nsp16 after collisional dissociation. The metal migration can be suppressed by using surface collision with reduced precursor charge states, which presumably resulted in minimal gas-phase structural rearrangement and highlighted the importance of complementary techniques. With minimal sample input (∼µg), native MS can rapidly detect ligand binding affinities and locations in dynamic multisubunit protein complexes, demonstrating the potential of an "all-in-one" native MS assay for rapid structural profiling of protein-to-AI-based compound systems to expedite drug discovery.


Subject(s)
Mass Spectrometry , Methyltransferases , Protein Multimerization , SARS-CoV-2 , Viral Nonstructural Proteins , Viral Regulatory and Accessory Proteins , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , SARS-CoV-2/chemistry , Mass Spectrometry/methods , Allosteric Regulation , Protein Binding , Humans , Ligands , Models, Molecular
14.
Cell Rep ; 43(3): 113941, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38478523

ABSTRACT

Resting CD4 T cells resist productive HIV-1 infection. The HIV-2/simian immunodeficiency virus protein viral accessory protein X (Vpx) renders these cells permissive to infection, presumably by alleviating blocks at cytoplasmic reverse transcription and subsequent nuclear import of reverse-transcription/pre-integration complexes (RTC/PICs). Here, spatial analyses using quantitative virus imaging techniques reveal that HIV-1 capsids containing RTC/PICs are readily imported into the nucleus, recruit the host dependency factor CPSF6, and translocate to nuclear speckles in resting CD4 T cells. Reverse transcription, however, remains incomplete, impeding proviral integration and viral gene expression. Vpx or pharmacological inhibition of the deoxynucleotide triphosphohydrolase (dNTPase) activity of the restriction factor SAM domain and HD domain-containing protein 1 (SAMHD1) increases levels of nuclear reverse-transcribed cDNA and facilitates HIV-1 integration. Nuclear import and intranuclear transport of viral complexes therefore do not pose important blocks to HIV-1 in resting CD4 T cells, and the limitation to reverse transcription by SAMHD1's dNTPase activity constitutes the main pre-integration block to infection.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Monomeric GTP-Binding Proteins , Animals , Humans , HIV-1/genetics , CD4-Positive T-Lymphocytes/metabolism , SAM Domain and HD Domain-Containing Protein 1/metabolism , HIV-2/genetics , Viral Regulatory and Accessory Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , HEK293 Cells
16.
Biochemistry ; 63(5): 632-643, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38377677

ABSTRACT

Hepatitis B virus X protein (HBx) plays a crucial role in the development of hepatocellular carcinoma (HCC) associated with hepatitis B virus (HBV) infection. The full-length HBx protein interacts with Bcl-xL and is involved in the HBV replication and cell death processes. The three hydrophobic residues Trp120, Leu123, and Ile127 of the HBx BH3-like motif are essential for the Bcl-xL-binding. On the other hand, various lengths of C-terminal-truncated HBx mutants are frequently detected in HCC tissues, and these mutants, rather than the full-length HBx, appear to be responsible for HCC development. Notably, the region spanning residues 1-120 of HBx [HBx(1 and 120)] has been strongly associated with an increased risk of HCC development. However, the mode of interaction between HBx(1-120) and Bcl-xL remains unclear. HBx(1-120) possesses only Trp120 among the three hydrophobic residues essential for the Bcl-xL-binding. To elucidate this interaction mode, we employed a C-terminal-deleted HBx BH3-like motif peptide composed of residues 101-120. Here, we present the NMR complex structure of Bcl-xL and HBx(101-120). Our results demonstrate that HBx(101-120) binds to Bcl-xL in a weaker manner. Considering the high expression of Bcl-xL in HCC cells, this weak interaction, in conjunction with the overexpression of Bcl-xL in HCC cells, may potentially contribute to HCC development through the interaction between C-terminal-truncated HBx and Bcl-xL.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Trans-Activators/chemistry , Viral Regulatory and Accessory Proteins/metabolism , bcl-X Protein/chemistry , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B/complications , Hepatitis B/pathology
17.
PLoS Biol ; 22(2): e3002544, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38422166

ABSTRACT

Ebolavirus (EBOV) belongs to a family of highly pathogenic viruses that cause severe hemorrhagic fever in humans. EBOV replication requires the activity of the viral polymerase complex, which includes the cofactor and Interferon antagonist VP35. We previously showed that the covalent ubiquitination of VP35 promotes virus replication by regulating interactions with the polymerase complex. In addition, VP35 can also interact non-covalently with ubiquitin (Ub); however, the function of this interaction is unknown. Here, we report that VP35 interacts with free (unanchored) K63-linked polyUb chains. Ectopic expression of Isopeptidase T (USP5), which is known to degrade unanchored polyUb chains, reduced VP35 association with Ub and correlated with diminished polymerase activity in a minigenome assay. Using computational methods, we modeled the VP35-Ub non-covalent interacting complex, identified the VP35-Ub interacting surface, and tested mutations to validate the interface. Docking simulations identified chemical compounds that can block VP35-Ub interactions leading to reduced viral polymerase activity. Treatment with the compounds reduced replication of infectious EBOV in cells and in vivo in a mouse model. In conclusion, we identified a novel role of unanchored polyUb in regulating Ebola virus polymerase function and discovered compounds that have promising anti-Ebola virus activity.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Nucleocapsid Proteins , Humans , Animals , Mice , Viral Regulatory and Accessory Proteins , Ubiquitin , Virus Replication , Ebolavirus/genetics
18.
J Mol Biol ; 436(4): 168423, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38185325

ABSTRACT

In bacteriophage λ lysogens, the λcI repressor is encoded by the leaderless transcript (lmRNA) initiated at the λpRM promoter. Translation is enhanced in rpsB mutants deficient in ribosomal protein uS2. Although translation initiation of lmRNA is conserved in bacteria, archaea, and eukaryotes, structural insight of a lmRNA translation initiation complex is missing. Here, we use cryo-EM to solve the structures of the uS2-deficient 70S ribosome of host E. coli mutant rpsB11 and the wild-type 70S complex with λcI lmRNA and fMet-tRNAfMet. Importantly, the uS2-deficient 70S ribosome also lacks protein bS21. The anti-Shine-Dalgarno (aSD) region is structurally supported by bS21, so that the absence of the latter causes the aSD to divert from the normal mRNA exit pathway, easing the exit of lmRNA. A π-stacking interaction between the monitor base A1493 and A(+4) of lmRNA potentially acts as a recognition signal. Coulomb charge flow, along with peristalsis-like dynamics within the mRNA entrance channel due to the increased 30S head rotation caused by the absence of uS2, are likely to facilitate the propagation of lmRNA through the ribosome. These findings lay the groundwork for future research on the mechanism of translation and the co-evolution of lmRNA and mRNA that includes the emergence of a defined ribosome-binding site of the transcript.


Subject(s)
Bacteriophage lambda , Escherichia coli , Peptide Chain Initiation, Translational , RNA, Messenger , Repressor Proteins , Ribosome Subunits, Large, Bacterial , Viral Regulatory and Accessory Proteins , Escherichia coli/genetics , Escherichia coli/virology , Ribosomal Proteins/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Bacteriophage lambda/genetics , Bacteriophage lambda/metabolism , Ribosome Subunits, Large, Bacterial/chemistry , Ribosome Subunits, Large, Bacterial/metabolism , Repressor Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics
19.
Virus Res ; 341: 199326, 2024 03.
Article in English | MEDLINE | ID: mdl-38253259

ABSTRACT

BACKGROUND: PreS1-binding protein (PreS1BP), recognized as a nucleolar protein and tumor suppressor, influences the replication of various viruses, including vesicular stomatitis virus (VSV) and herpes simplex virus type 1 (HSV-1). Its role in hepatitis B virus (HBV) replication and the underlying mechanisms, however, remain elusive. METHODS: We investigated PreS1BP expression levels in an HBV-replicating cell and animal model and analyzed the impact of its overexpression on viral replication metrics. HBV DNA, covalently closed circular DNA (cccDNA), hepatitis B surface antigen (HBsAg), hepatitis B core antigen (HBcAg), and HBV RNA levels were assessed in HBV-expressing stable cell lines under varying PreS1BP conditions. Furthermore, co-immunoprecipitation and ubiquitination assays were used to detect PreS1BP- hepatitis B virus X protein (HBx) interactions and HBx stability modulated by PreS1BP. RESULTS: Our study revealed a marked decrease in PreS1BP expression in the presence of active HBV replication. Functional assays showed that PreS1BP overexpression significantly inhibited HBV replication and transcription, evidenced by the reduction in HBV DNA, cccDNA, HBsAg, HBcAg, and HBV RNA levels. At the molecular level, PreS1BP facilitated the degradation of HBx in a dose-dependent fashion, whereas siRNA-mediated knockdown of PreS1BP led to an increase in HBx levels. Subsequent investigations uncovered that PreS1BP accelerated HBx protein degradation via K63-linked ubiquitination in a ubiquitin-proteasome system-dependent manner. Co-immunoprecipitation assays further established that PreS1BP enhances the recruitment of the proteasome 20S subunit alpha 3 (PSMA3) for interaction with HBx, thereby fostering its degradation. CONCLUSIONS: These findings unveil a previously unidentified mechanism wherein PreS1BP mediates HBx protein degradation through the ubiquitin-proteasome system, consequentially inhibiting HBV replication. This insight positions PreS1BP as a promising therapeutic target for future HBV interventions. Further studies are warranted to explore the clinical applicability of modulating PreS1BP in HBV therapy.


Subject(s)
Hepatitis B virus , Hepatitis B , Animals , Humans , Hepatitis B virus/genetics , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/metabolism , Proteolysis , Hepatitis B Core Antigens/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Proteasome Endopeptidase Complex/metabolism , Hep G2 Cells , Viral Regulatory and Accessory Proteins/genetics , DNA, Circular/metabolism , Virus Replication/genetics , RNA/metabolism , Ubiquitins/genetics
20.
Biochemistry ; 63(3): 241-250, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38216552

ABSTRACT

Viroporins constitute a class of viral membrane proteins with diverse roles in the viral life cycle. They can self-assemble and form pores within the bilayer that transport substrates, such as ions and genetic material, that are critical to the viral infection cycle. However, there is little known about the oligomeric state of most viroporins. Here, we use native mass spectrometry in detergent micelles to uncover the patterns of oligomerization of the full-length SARS-CoV-2 envelope (E) protein, poliovirus VP4, and HIV Vpu. Our data suggest that the E protein is a specific dimer, VP4 is exclusively monomeric, and Vpu assembles into a polydisperse mixture of oligomers under these conditions. Overall, these results revealed the diversity in the oligomerization of viroporins, which has implications for the mechanisms of their biological functions as well as their potential as therapeutic targets.


Subject(s)
COVID-19 , HIV Infections , Poliovirus , Humans , SARS-CoV-2/metabolism , Viroporin Proteins , Viral Regulatory and Accessory Proteins , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...