Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.466
Filter
1.
Proc Natl Acad Sci U S A ; 121(24): e2400378121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38830096

ABSTRACT

Epitranscriptomic RNA modifications have emerged as important regulators of the fate and function of viral RNAs. One prominent modification, the cytidine methylation 5-methylcytidine (m5C), is found on the RNA of HIV-1, where m5C enhances the translation of HIV-1 RNA. However, whether m5C functionally enhances the RNA of other pathogenic viruses remains elusive. Here, we surveyed a panel of commonly found RNA modifications on the RNA of hepatitis B virus (HBV) and found that HBV RNA is enriched with m5C as well as ten other modifications, at stoichiometries much higher than host messenger RNA (mRNA). Intriguingly, m5C is mostly found on the epsilon hairpin, an RNA element required for viral RNA encapsidation and reverse transcription, with these m5C mainly deposited by the cellular methyltransferase NSUN2. Loss of m5C from HBV RNA due to NSUN2 depletion resulted in a partial decrease in viral core protein (HBc) production, accompanied by a near-complete loss of the reverse transcribed viral DNA. Similarly, mutations introduced to remove the methylated cytidines resulted in a loss of HBc production and reverse transcription. Furthermore, pharmacological disruption of m5C deposition led to a significant decrease in HBV replication. Thus, our data indicate m5C methylations as a critical mediator of the epsilon elements' function in HBV virion production and reverse transcription, suggesting the therapeutic potential of targeting the m5C methyltransfer process on HBV epsilon as an antiviral strategy.


Subject(s)
Cytidine , Hepatitis B virus , RNA, Viral , Reverse Transcription , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , Cytidine/analogs & derivatives , Cytidine/metabolism , Cytidine/genetics , Humans , Reverse Transcription/genetics , Methylation , Virus Replication/genetics , Epigenesis, Genetic , Virion/metabolism , Virion/genetics , Transcriptome
2.
J Int Med Res ; 52(5): 3000605241252580, 2024 May.
Article in English | MEDLINE | ID: mdl-38760056

ABSTRACT

Recombinant human type II tumour necrosis factor receptor-antibody fusion protein (rh TNFR:Fc) is an immunosuppressant approved for treating rheumatoid arthritis (RA). This case report describes a case of hepatitis B reactivation in a patient with drug-induced acute-on-chronic liver failure. A 58-year-old woman with a history of RA was treated with rh TNFR:Fc; and then subsequently received 25 mg rh TNFR:Fc, twice a week, as maintenance therapy. No anti-hepatitis B virus (HBV) preventive treatment was administered. Six months later, she was hospitalized with acute jaundice. HBV reactivation was observed, leading to acute-on-chronic liver failure. After active treatment, the patient's condition improved and she recovered well. Following careful diagnosis and treatment protocols are essential when treating RA with rh TNFR:Fc, especially in anti-hepatitis B core antigen antibody-positive patients, even when the HBV surface antigen and the HBV DNA are negative. In the case of HBV reactivation, liver function parameters, HBV surface antigen and HBV DNA should be closely monitored during treatment, and antiviral drugs should be used prophylactically when necessary, as fatal hepatitis B reactivation may occur in rare cases. A comprehensive evaluation and medication should be administered in a timely manner after evaluating the patient's physical condition and closely monitoring the patient.


Subject(s)
Hepatitis B virus , Hepatitis B , Recombinant Fusion Proteins , Virus Activation , Humans , Female , Middle Aged , Hepatitis B virus/pathogenicity , Hepatitis B virus/physiology , Virus Activation/drug effects , Recombinant Fusion Proteins/therapeutic use , Hepatitis B/virology , Hepatitis B/drug therapy , Hepatitis B/complications , Hepatitis B/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/virology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/complications , Liver Failure/virology , Liver Failure/etiology
3.
Biol Pharm Bull ; 47(5): 941-945, 2024.
Article in English | MEDLINE | ID: mdl-38735754

ABSTRACT

Hepatitis B virus reactivation (HBV-R) is a serious complication that can occur in patients with resolved HBV infection during cancer chemotherapy. We examined the levels of HBV surface antibody (HBsAb) and HBV core antibody (HBcAb) to assess the incidence of HBV-R in cancer patients including hematopoietic stem cell transplantation (HSCT) and rituximab administration. This retrospective cohort study included 590 patients with resolved HBV infection. The incidence of HBV-R was evaluated 761.5 (range, 90-3898) days after the inititiation of chemotherapy. Of the patients, 13 (2.2%) developed HBV-R after the start of chemotherapy. All 13 patients exhibited lower HBsAb (<100 mIU/mL) levels at baseline. A higher level of HBcAb (≥100 cut off index (C.O.I.)) was a possible risk factor for HBV-R as well as HSCT and rituximab administration. The simultaneous presence of HBsAb <100 mIU/mL and HBcAb ≥100 C.O.I. increased the risk of HBV-R by 18.5%. Patients treated with rituximab were at a higher risk of HBV-R (18.4%) despite having HBcAb <100 C.O.I. Our results suggest that assessment of HBsAb and HBcAb levels prior to the chemotherapy is important for identifying patients at high risk of HBV-R, especially in solid cancers without HSCT and rituximab administration.


Subject(s)
Hepatitis B Antibodies , Hepatitis B Surface Antigens , Hepatitis B virus , Hepatitis B , Rituximab , Virus Activation , Humans , Male , Female , Middle Aged , Retrospective Studies , Hepatitis B Antibodies/blood , Hepatitis B Antibodies/immunology , Hepatitis B virus/immunology , Hepatitis B virus/physiology , Virus Activation/drug effects , Rituximab/therapeutic use , Rituximab/adverse effects , Adult , Aged , Hepatitis B/immunology , Hepatitis B Surface Antigens/blood , Hepatitis B Surface Antigens/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Young Adult , Neoplasms/drug therapy , Neoplasms/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/blood , Aged, 80 and over , Adolescent
4.
Cell Commun Signal ; 22(1): 280, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773448

ABSTRACT

INTRODUCTION: Hepatitis B Virus (HBV) is widely recognized as a "metabolic virus" that disrupts hepatic metabolic homeostasis, rendering it one of the foremost risk factors for hepatocellular carcinoma (HCC). Except for antiviral therapy, the fundamental principles underlying HBV- and HBV+ HCC have remained unchanged, limiting HCC treatment options. OBJECTIVES: In this study, we aim to identify the distinctive metabolic profile of HBV-associated HCC, with the promise of identifying novel metabolic targets that confer survival advantages and ultimately impede cancer progression. METHODS: We employed a comprehensive methodology to evaluate metabolic alterations systematically. Initially, we analyzed transcriptomic and proteomic data obtained from a public database, subsequently validating these findings within our test cohort at both the proteomic and transcriptomic levels. Additionally, we conducted a comprehensive analysis of tissue metabolomics profiles, lipidomics, and the activity of the MAPK and AKT signaling pathway to corroborate the abovementioned changes. RESULTS: Our multi-omics approach revealed distinct metabolic dysfunctions associated with HBV-associated HCC. Specifically, we observed upregulated steroid hormone biosynthesis, primary bile acid metabolism, and sphingolipid metabolism in HBV-associated HCC patients' serum. Notably, metabolites involved in primary bile acid and sphingolipids can activate the MAPK/mTOR pathway. Tissue metabolomics and lipidomics analyses further validated the serum metabolic alterations, particularly alterations in lipid composition and accumulation of unsaturated fatty acids. CONCLUSION: Our findings emphasize the pivotal role of HBV in HCC metabolism, elucidating the activation of a unique MAPK/mTOR signaling axis by primary bile acids and sphingolipids. Moreover, the hyperactive MAPK/mTOR signaling axis transduction leads to significant reprogramming in lipid metabolism within HCC cells, further triggering the activation of the MAPK/mTOR pathway in turn, thereby establishing a self-feeding circle driven by primary bile acids and sphingolipids.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B virus , Liver Neoplasms , TOR Serine-Threonine Kinases , Humans , Carcinoma, Hepatocellular/virology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/virology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Hepatitis B virus/physiology , Lipid Metabolism , Male , Lipids/blood , Signal Transduction , MAP Kinase Signaling System , Hepatitis B/complications , Hepatitis B/virology , Hepatitis B/metabolism , Middle Aged , Female
5.
J Immunol Res ; 2024: 4722047, 2024.
Article in English | MEDLINE | ID: mdl-38745751

ABSTRACT

Hepatitis B virus (HBV) infection is a major global health issue and ranks among the top causes of liver cirrhosis and hepatocellular carcinoma. Although current antiviral medications, including nucleot(s)ide analogs and interferons, could inhibit the replication of HBV and alleviate the disease, HBV cannot be fully eradicated. The development of cellular and animal models for HBV infection plays an important role in exploring effective anti-HBV medicine. During the past decades, advancements in several cell culture systems, such as HepG2.2.15, HepAD38, HepaRG, hepatocyte-like cells, and primary human hepatocytes, have propelled the research in inhibiting HBV replication and expression and thus enriched our comprehension of the viral life cycle and enhancing antiviral drug evaluation efficacy. Mouse models, in particular, have emerged as the most extensively studied HBV animal models. Additionally, the present landscape of HBV therapeutics research now encompasses a comprehensive assessment of the virus's life cycle, targeting numerous facets and employing a variety of immunomodulatory approaches, including entry inhibitors, strategies aimed at cccDNA, RNA interference technologies, toll-like receptor agonists, and, notably, traditional Chinese medicine (TCM). This review describes the attributes and limitations of existing HBV model systems and surveys novel advancements in HBV treatment modalities, which will offer deeper insights toward discovering potentially efficacious pharmaceutical interventions.


Subject(s)
Antiviral Agents , Disease Models, Animal , Hepatitis B virus , Hepatitis B , Virus Replication , Humans , Animals , Hepatitis B virus/physiology , Hepatitis B virus/drug effects , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Hepatitis B/drug therapy , Hepatitis B/virology , Hepatitis B/immunology , Virus Replication/drug effects , Mice , Hepatocytes/virology
6.
Viruses ; 16(5)2024 04 30.
Article in English | MEDLINE | ID: mdl-38793598

ABSTRACT

Chronic Hepatitis B and D Virus (HBV and HDV) co-infection is responsible for the most severe form of viral Hepatitis, the Hepatitis Delta. Despite an efficient vaccine against HBV, the HBV/HDV infection remains a global health burden. Notably, no efficient curative treatment exists against any of these viruses. While physiologically distinct, HBV and HDV life cycles are closely linked. HDV is a deficient virus that relies on HBV to fulfil is viral cycle. As a result, the cellular response to HDV also influences HBV replication. In vitro studying of HBV and HDV infection and co-infection rely on various cell culture models that differ greatly in terms of biological relevance and amenability to classical virology experiments. Here, we review the various cell culture models available to scientists to decipher HBV and HDV virology and host-pathogen interactions. We discuss their relevance and how they may help address the remaining questions, with one objective in mind: the development of new therapeutic approaches allowing viral clearance in patients.


Subject(s)
Hepatitis B virus , Hepatitis D , Hepatitis Delta Virus , Virus Replication , Humans , Hepatitis Delta Virus/physiology , Hepatitis Delta Virus/genetics , Hepatitis B virus/physiology , Hepatitis D/virology , Animals , Host-Pathogen Interactions , Coinfection/virology , Cell Culture Techniques , Hepatitis B/virology
7.
Viruses ; 16(5)2024 05 03.
Article in English | MEDLINE | ID: mdl-38793606

ABSTRACT

Hepatitis B and C viruses (HBV and HCV) are the leading causes of end-stage liver disease worldwide. Although there is a potent vaccine against HBV, many new infections are recorded annually, especially in poorly resourced places which have lax vaccination policies. Again, as HBV has no cure and chronic infection is lifelong, vaccines cannot help those already infected. Studies to thoroughly understand the HBV biology and pathogenesis are limited, leaving much yet to be understood about the genomic features and their role in establishing and maintaining infection. The current knowledge of the impact on disease progression and response to treatment, especially in hyperendemic regions, is inadequate. This calls for in-depth studies on viral biology, mainly for the purposes of coming up with better management strategies for infected people and more effective preventative measures for others. This information could also point us in the direction of a cure. Here, we discuss the progress made in understanding the genomic basis of viral activities leading to the complex interplay of the virus and the host, which determines the outcome of HBV infection as well as the impact of coinfections.


Subject(s)
Hepatitis B virus , Hepatitis B , Humans , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B/virology , Coinfection/virology , Genome, Viral , Animals
8.
Viruses ; 16(5)2024 05 08.
Article in English | MEDLINE | ID: mdl-38793623

ABSTRACT

Hepatitis B virus (HBV) is a major driver of chronic hepatic inflammation, which regularly leads to liver cirrhosis or hepatocellular carcinoma. Immediate innate immune cell response is crucial for the rapid clearance of the infection. Here, natural killer (NK) cells play a pivotal role in direct cytotoxicity and the secretion of antiviral cytokines as well as regulatory function. The aim of this study was to further elucidate NK cell responses triggered by an HBV infection. Therefore, we optimized HBV in vitro models that reliably stimulate NK cells using hepatocyte-like HepG2 cells expressing the Na+-taurocholate co-transporting polypeptide (NTCP) and HepaRG cells. Immune cells were acquired from healthy platelet donors. Initially, HepG2-NTCP cells demonstrated higher viral replication compared to HepaRG cells. Co-cultures with immune cells revealed increased production of interferon-γ and tumor necrosis factor-α by NK cells, which was no longer evident in isolated NK cells. Likewise, the depletion of monocytes and spatial separation from target cells led to the absence of the antiviral cytokine production of NK cells. Eventually, the combined co-culture of isolated NK cells and monocytes led to a sufficient cytokine response of NK cells, which was also apparent when communication between the two immune cell subpopulations was restricted to soluble factors. In summary, our study demonstrates antiviral cytokine production by NK cells in response to HBV+ HepG2-NTCP cells, which is dependent on monocyte bystander activation.


Subject(s)
Coculture Techniques , Cytokines , Hepatitis B virus , Hepatitis B , Killer Cells, Natural , Monocytes , Humans , Killer Cells, Natural/immunology , Monocytes/immunology , Monocytes/virology , Hepatitis B virus/immunology , Hepatitis B virus/physiology , Cytokines/metabolism , Hep G2 Cells , Hepatitis B/immunology , Hepatitis B/virology , Virus Replication , Interferon-gamma/metabolism , Interferon-gamma/immunology , Tumor Necrosis Factor-alpha/metabolism , Hepatocytes/virology , Hepatocytes/immunology
9.
Viruses ; 16(5)2024 05 11.
Article in English | MEDLINE | ID: mdl-38793645

ABSTRACT

The hepatitis B virus (HBV) infects hepatocytes and hijacks host cellular mechanisms for its replication. Host proteins can be frontline effectors of the cell's defense and restrict viral replication by impeding multiple steps during its intracellular lifecycle. This review summarizes many of the well-described restriction factors, their mechanisms of restriction, and counteractive measures of HBV, with a special focus on viral transcription. We discuss some of the limitations and knowledge gaps about the restriction factors, highlighting how these factors may be harnessed to facilitate therapeutic strategies against HBV.


Subject(s)
Hepatitis B virus , Hepatitis B , Hepatocytes , Host-Pathogen Interactions , Virus Replication , Hepatitis B virus/physiology , Hepatitis B virus/genetics , Humans , Hepatitis B/virology , Hepatocytes/virology , Animals
10.
Int Immunopharmacol ; 134: 112219, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38733823

ABSTRACT

BACKGROUNDS & AIMS: Given its ability to inhibit HBV replication, Interferon alpha (IFN-α) treatment has been confirmed to be effective in managing Chronic Hepatitis B (CHB). However, its underlying mechanisms are incompletely understood. METHODS: Herein, we investigated the antiviral properties of IFN-α by introducing IFN-α expression plasmids into a well-established HBV Hydrodynamic Injection (HDI) mouse model and examined the impact of IFN-α or hepcidin treatment on macrophages derived from THP-1 cells. The cytokine profiles were analyzed using the cytometry microsphere microarray technology, and flow cytometry was used to analyze the polarization of macrophages. Additionally, the IL-6/JAK2/STAT3 signaling pathway and the hepcidin-ferroportin axis were analyzed to better understand the macrophage polarization mechanism. RESULTS: As evidenced by the suppression of HBV replication, injection of an IFN-α expression plasmid and supernatants of IFN-α-treated macrophages exerted anti-HBV effects. The IFN-α treatment up-regulated IL-6 in mice with HBV replication, as well as in IFN-α-treated HepG2 cells and macrophages. Furthermore, JAK2/STAT3 signaling and hepcidin expression was promoted, inducing iron accumulation via the hepcidin-ferroportin axis, which caused the polarization of M1 macrophages. Furthermore, under the effect of IFN-α, IL-6 silencing or blockade downregulated the JAK2/STAT3 signaling pathway and hepcidin, implying that increased hepcidin expression under IFN-α treatment was dependent on the IL-6/JAK2/STAT3 pathway. CONCLUSION: The IL-6/JAK2/STAT3 signaling pathway is activated by IFN-α which induces hepcidin expression. The resulting iron accumulation then induces the polarization of M1 macrophages via the hepcidin-ferroportin axis, yielding an immune response which exerts antiviral effects against HBV replication.


Subject(s)
Antiviral Agents , Hepatitis B virus , Hepcidins , Interferon-alpha , Janus Kinase 2 , Macrophages , STAT3 Transcription Factor , Hepcidins/metabolism , Hepcidins/genetics , Animals , Humans , Interferon-alpha/pharmacology , Macrophages/immunology , Macrophages/drug effects , Hepatitis B virus/physiology , Hepatitis B virus/drug effects , Hepatitis B virus/immunology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Mice , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Hep G2 Cells , Signal Transduction/drug effects , Interleukin-6/metabolism , THP-1 Cells , Mice, Inbred C57BL , Virus Replication/drug effects , Male , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Disease Models, Animal , Hepatitis B/immunology , Hepatitis B/drug therapy , Hepatitis B/virology , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics
12.
J Med Virol ; 96(5): e29659, 2024 May.
Article in English | MEDLINE | ID: mdl-38747016

ABSTRACT

Hepatitis B virus (HBV) infection is a major global health burden with 820 000 deaths per year. In our previous study, we found that the knockdown of autophagy-related protein 5 (ATG5) significantly upregulated the interferon-stimulated genes (ISGs) expression to exert the anti-HCV effect. However, the regulation of ATG5 on HBV replication and its underlying mechanism remains unclear. In this study, we screened the altered expression of type I interferon (IFN-I) pathway genes using RT² Profiler™ PCR array following ATG5 knock-down and we found the bone marrow stromal cell antigen 2 (BST2) expression was significantly increased. We then verified the upregulation of BST2 by ATG5 knockdown using RT-qPCR and found that the knockdown of ATG5 activated the Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling pathway. ATG5 knockdown or BST2 overexpression decreased Hepatitis B core Antigen (HBcAg) protein, HBV DNA levels in cells and supernatants of HepAD38 and HBV-infected NTCP-HepG2. Knockdown of BST2 abrogated the anti-HBV effect of ATG5 knockdown. Furthermore, we found that ATG5 interacted with BST2, and further formed a ternary complex together with HBV-X (HBx). In conclusion, our finding indicates that ATG5 promotes HBV replication through decreasing BST2 expression and interacting with it directly to antagonize its antiviral function.


Subject(s)
Antigens, CD , Autophagy-Related Protein 5 , Bone Marrow Stromal Antigen 2 , GPI-Linked Proteins , Hepatitis B virus , Virus Replication , Humans , Antigens, CD/genetics , Antigens, CD/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Gene Knockdown Techniques , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/genetics , Hep G2 Cells , Hepatitis B/virology , Hepatitis B/genetics , Hepatitis B virus/physiology , Hepatitis B virus/genetics , Host-Pathogen Interactions , Signal Transduction , Bone Marrow Stromal Antigen 2/metabolism
13.
J Gen Virol ; 105(5)2024 05.
Article in English | MEDLINE | ID: mdl-38757942

ABSTRACT

Since its discovery in 1965, our understanding of the hepatitis B virus (HBV) replication cycle and host immune responses has increased markedly. In contrast, our knowledge of the molecular biology of hepatitis delta virus (HDV), which is associated with more severe liver disease, is less well understood. Despite the progress made, critical gaps remain in our knowledge of HBV and HDV replication and the mechanisms underlying viral persistence and evasion of host immunity. The International HBV Meeting is the leading annual scientific meeting for presenting the latest advances in HBV and HDV molecular virology, immunology, and epidemiology. In 2023, the annual scientific meeting was held in Kobe, Japan and this review summarises some of the advances presented at the Meeting and lists gaps in our knowledge that may facilitate the development of new therapies.


Subject(s)
Hepatitis B virus , Hepatitis B , Hepatitis Delta Virus , Virus Replication , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B virus/immunology , Humans , Hepatitis Delta Virus/genetics , Hepatitis Delta Virus/physiology , Hepatitis B/virology , Hepatitis B/immunology , Molecular Biology , Japan , Hepatitis D/virology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics
14.
Immunobiology ; 229(3): 152810, 2024 May.
Article in English | MEDLINE | ID: mdl-38772101

ABSTRACT

BACKGROUND AND AIMS: Activation of the cGAS-STING pathway induces the production of type I interferons, initiating the antiviral immune response, which contributes to the clearance of pathogens. Previous studies have shown that STING agonists promote hepatitis B virus (HBV) clearance; however, few studies have investigated the effect of activating the cGAS-STING pathway in macrophages on HBV. METHODS: The polarization status of HBV particle-stimulated RAW264.7 macrophages was analyzed. After stimulation with HBV particles, the analysis focused on determining whether the DNA sensors in RAW264.7 macrophages recognized the viral double-stranded DNA (dsDNA) and evaluating the activation of the cGAS-STING pathway. Coculture of mouse macrophages and hepatocytes harboring HBV was used to study the antiviral activity of HBV-stimulated RAW264.7 macrophages. RESULTS: After stimulation with HBV particles, HBV relaxed circular DNA (rcDNA) was detected in RAW264.7 macrophages, and the protein expression of phospho-STING, phospho-TBK1, and phospho-IRF3 in the STING pathway was increased, as shown by Western blot analysis, which revealed that M1 polarization of macrophages was caused by increased expression of CD86. RT-PCR analyses revealed elevated expression of M1 macrophage polarization-associated cytokines such as TNFα, IL-1ß, iNOS, and IFNα/ß. In the coculture experiment, both HBsAg and HBeAg expression levels were significantly decreased in AML12-HBV1.3 cells cocultured with the supernatants of HBV-stimulated RAW264.7 macrophages. CONCLUSION: The results suggest that macrophages can endocytose HBV particles. Additionally, viral dsDNA can be recognized by DNA pattern recognition receptors, which in turn activate the cGAS-STING pathway, promoting the M1 polarization of macrophages, while no significant M2 polarization is observed. Macrophages stimulated with HBV particles exhibit enhanced antiviral activity against HBV.


Subject(s)
DNA, Viral , Hepatitis B virus , Macrophages , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Hepatitis B virus/physiology , Hepatitis B virus/immunology , Animals , Nucleotidyltransferases/metabolism , Mice , Macrophages/immunology , Macrophages/virology , Macrophages/metabolism , Membrane Proteins/metabolism , RAW 264.7 Cells , Hepatitis B/immunology , Hepatitis B/virology , Humans , Macrophage Activation/immunology , Hepatocytes/virology , Hepatocytes/immunology , Hepatocytes/metabolism , Interferon Regulatory Factor-3/metabolism
15.
Viruses ; 16(5)2024 05 08.
Article in English | MEDLINE | ID: mdl-38793626

ABSTRACT

HBV infection is challenging to cure due to the persistence of viral covalently closed circular viral DNA (cccDNA). The dedicator of cytokinesis 11 (DOCK11) is recognized as a guanine nucleotide exchange factor (GEF) for CDC42 that has been reported to be required for HBV persistence. DOCK11 is expressed in both the cytoplasm and nucleus of human hepatocytes and is functionally associated with retrograde trafficking proteins Arf-GAP with GTPase domain, ankyrin repeat, and pleckstrin homology domain-containing protein 2 (AGAP2), and ADP-ribosylation factor 1 (ARF1), together with the HBV capsid, in the trans-Golgi network (TGN). This opens an alternative retrograde trafficking route for HBV from early endosomes (EEs) to the TGN and then to the endoplasmic reticulum (ER), thereby avoiding lysosomal degradation. DOCK11 also facilitates the association of cccDNA with H3K4me3 and RNA Pol II for activating cccDNA transcription. In addition, DOCK11 plays a crucial role in the host DNA repair system, being essential for cccDNA synthesis. This function can be inhibited by 10M-D42AN, a novel DOCK11-binding peptide, leading to the suppression of HBV replication both in vitro and in vivo. Treatment with a combination of 10M-D42AN and entecavir may represent a promising therapeutic strategy for patients with chronic hepatitis B (CHB). Consequently, DOCK11 may be seen as a potential candidate molecule in the development of molecularly targeted drugs against CHB.


Subject(s)
Guanine Nucleotide Exchange Factors , Hepatitis B virus , Hepatocytes , Humans , Hepatitis B virus/physiology , Hepatitis B virus/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Hepatocytes/virology , Hepatocytes/metabolism , Virus Internalization , Virus Replication , Hepatitis B/virology , Hepatitis B/metabolism , DNA, Viral/metabolism , DNA, Viral/genetics , Animals
16.
Front Immunol ; 15: 1330644, 2024.
Article in English | MEDLINE | ID: mdl-38558804

ABSTRACT

Background: Previous studies have suggested the potential of PD-1/PD-L1 inhibitors in the treatment of chronic HBV infection. However, since phase III clinical trials have not yet been announced, additional clinical insights may be obtained by observing changes in serum hepatitis B surface antigen (HBsAg) and HBV-DNA levels in cancer patients undergoing PD-1 inhibitor therapy. Objective: To explore the effects of PD-1 inhibitor combinational therapy on serum HBsAg and HBV-DNA levels, investigate the incidence of HBsAg loss, HBV reactivation (HBVr), and immune-related adverse events (irAEs), and identify the risk factors associated with significant HBsAg fluctuations and HBVr. Methods: A retrospective study including 1195 HBsAg-positive cancer patients who received PD-1 inhibitors between July 2019 and June 2023 was conducted, and 180 patients were enrolled in this study. Serum HBsAg levels before and after PD-1 inhibitor administration were compared across different subgroups. The Pearson χ2 or Fisher exact test was performed to investigate the relationships between categorical variables. Univariable and multivariable analysis were performed to identify the risk factors associated with significant HBsAg fluctuations and HBVr. Results: With the concurrent use of antiviral agents, serum HBsAg levels decreased (Z=-3.966, P < 0.0001) in 129 patients and increased (t=-2.047, P=0.043) in 51 patients. Additionally, 7 patients (3.89%) achieved serum HBsAg loss. Virus replication was suppressed in most of the enrolled patients. When divided patients into different subgroups, significant HBsAg decreases after PD-1 inhibitor administration were discovered in lower baseline HBsAg group (Z=-2.277, P=0.023), HBeAg-seronegative group (Z=-2.200, P=0.028), non-irAEs occurrence group (Z=-2.007, P=0.045) and liver cancer group (Z=-1.987, P=0.047). Of note, 11 patients and 36 patients experienced HBVr (6.11%) and irAEs (20%), respectively, which could lead to discontinuation or delayed use of PD-1 inhibitors. After multivariable analysis, HBeAg-seropositive (OR, 7.236 [95% CI, 1.757-29.793], P=0.01) and the occurrence of irAEs (OR, 4.077 [95% CI, 1.252-13.273], P=0.02) were identified as the independent risk factors for significant HBsAg increase, the occurrence of irAEs (OR, 5.560 [95% CI, 1.252-13.273], P=0.01) was identified as the only independent risk factor for HBVr. Conclusion: PD-1 inhibitors combined with nucleos(t)ide analogues (NAs) may exert therapeutic potential for chronic HBV infection in cancer patients. However, attention also should be paid to the risk of significant elevation in HBsAg levels, HBVr, and irAEs associated with PD-1 inhibitor combinational therapy.


Subject(s)
Hepatitis B Surface Antigens , Neoplasms , Humans , Hepatitis B virus/physiology , Immune Checkpoint Inhibitors/adverse effects , Hepatitis B e Antigens , Retrospective Studies , DNA, Viral , Risk Factors , Neoplasms/drug therapy
17.
Viruses ; 16(4)2024 03 29.
Article in English | MEDLINE | ID: mdl-38675875

ABSTRACT

Individuals chronically infected with hepatitis B virus (HBV) and hepatitis Delta virus (HDV) present an increased risk of developing cirrhosis and hepatocellular carcinoma in comparison to HBV mono-infected individuals. Although HDV only replicates in individuals coinfected or superinfected with HBV, there is currently no in vitro model that can stably express both viruses simultaneously, mimicking the chronic infections seen in HBV/HDV patients. Here, we present the HepG2BD cell line as a novel in vitro culture system for long-term replication of HBV and HDV. HepG2BD cells derive from HepG2.2.15 cells in which a 2 kb HDV cDNA sequence was inserted into the adeno-associated virus safe harbor integration site 1 (AAVS1) using CRISPR-Cas9. A Tet-Off promoter was placed 5' of the genomic HDV sequence for reliable initiation/repression of viral replication and secretion. HBV and HDV replication were then thoroughly characterized. Of note, non-dividing cells adopt a hepatocyte-like morphology associated with an increased production of both HDV and HBV virions. Finally, HDV seems to negatively interfere with HBV in this model system. Altogether, HepG2BD cells will be instrumental to evaluate, in vitro, the fundamental HBV-HDV interplay during simultaneous chronic replication as well as for antivirals screening targeting both viruses.


Subject(s)
Hepatitis B virus , Hepatitis Delta Virus , Virus Replication , Hepatitis Delta Virus/genetics , Hepatitis Delta Virus/physiology , Humans , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hep G2 Cells , Hepatocytes/virology , Hepatitis D/virology , CRISPR-Cas Systems , Dependovirus/genetics , Coinfection/virology
18.
Viruses ; 16(4)2024 03 29.
Article in English | MEDLINE | ID: mdl-38675871

ABSTRACT

The post-transcriptional regulatory element (PRE) is present in all HBV mRNAs and plays a major role in their stability, nuclear export, and enhancement of viral gene expression. Understanding PRE's structure, function, and mode of action is essential to leverage its potential as a therapeutic target. A wide range of PRE-based reagents and tools have been developed and assessed in preclinical and clinical settings for therapeutic and biotechnology applications. This manuscript aims to provide a systematic review of the characteristics and mechanism of action of PRE, as well as elucidating its current applications in basic and clinical research. Finally, we discuss the promising opportunities that PRE may provide to antiviral development, viral biology, and potentially beyond.


Subject(s)
Hepatitis B virus , Hepatitis B , RNA, Viral , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , Hepatitis B/virology , Hepatitis B/drug therapy , RNA, Viral/genetics , RNA, Viral/metabolism , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Gene Expression Regulation, Viral , RNA, Messenger/genetics , RNA Processing, Post-Transcriptional , Animals
19.
Viruses ; 16(4)2024 04 15.
Article in English | MEDLINE | ID: mdl-38675950

ABSTRACT

Hepatitis B virus (HBV) is the etiologic agent of chronic hepatitis B, which puts at least 300 million patients at risk of developing fibrosis, cirrhosis, and hepatocellular carcinoma. HBV is a partially double-stranded DNA virus of the Hepadnaviridae family. While HBV was discovered more than 50 years ago, many aspects of its replicative cycle remain incompletely understood. Central to HBV persistence is the formation of covalently closed circular DNA (cccDNA) from the incoming relaxed circular DNA (rcDNA) genome. cccDNA persists as a chromatinized minichromosome and is the major template for HBV gene transcription. Here, we review how cccDNA and the viral minichromosome are formed and how viral gene transcription is regulated and highlight open questions in this area of research.


Subject(s)
DNA, Circular , DNA, Viral , Hepatitis B virus , Virus Replication , Hepatitis B virus/genetics , Hepatitis B virus/physiology , DNA, Circular/genetics , Humans , DNA, Viral/genetics , Viral Transcription/genetics , Gene Expression Regulation, Viral , Transcription, Genetic , Genome, Viral , Hepatitis B, Chronic/virology , Hepatitis B/virology , DNA Replication
20.
Viruses ; 16(4)2024 04 16.
Article in English | MEDLINE | ID: mdl-38675959

ABSTRACT

Hepatitis B virus (HBV) infections affect approximately 296 million people around the world, and the prevalence of any past or present HBV infection during the years 2015-2018 was as high as 4.3%. Acute HBV infection often presents with nonspecific symptoms and is usually self-limited, but 5% of patients can have persistent infections leading to chronic HBV infection and the risk of turning into chronic HBV infection is significantly higher in babies with vertical transmission (95%). Patients with chronic HBV infection are usually asymptomatic, but 15 to 40% of chronic HBV carriers develop cirrhosis and/or hepatocellular carcinoma. In addition to liver-related disorders, HBV is also associated with several extrahepatic complications, including glomerulonephritis, cryoglobulinemia, neurologic disorders, psychological manifestations, polyarthritis, and dermatologic disorders. Making the diagnosis of HBV can be challenging since patients with chronic infections can remain symptom-free for decades before developing cirrhosis or hepatocellular carcinoma, and patients with acute HBV infection may have only mild, nonspecific symptoms. Therefore, understanding how this virus causes extrahepatic complications can help clinicians consider this possibility in patients with diverse symptom presentations. The pathophysiology of these extrahepatic disorders likely involves immune-related tissue injury following immune complex formation and inflammatory cascades. In some cases, direct viral infection of extrahepatic tissue may cause a clinical syndrome. Currently, the American Association for the Study of Liver Diseases recommends treatment of chronic HBV infections with interferon therapy and/or nucleos(t)ide analogs, and this treatment has been reported to improve some extrahepatic disorders in some patients with chronic HBV infection. These extrahepatic complications have a significant role in disease outcomes and increase medical costs, morbidity, and mortality. Therefore, understanding the frequency and pathogenesis of these extrahepatic complications provides important information for both specialists and nonspecialists and may help clinicians identify patients at an earlier stage of their infection.


Subject(s)
Comorbidity , Hepatitis B virus , Humans , Hepatitis B virus/physiology , Hepatitis B/epidemiology , Hepatitis B/complications , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/epidemiology , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/virology , Liver Neoplasms/epidemiology , Liver Neoplasms/virology , Liver Cirrhosis/epidemiology , Liver Cirrhosis/virology , Cost of Illness , Antiviral Agents/therapeutic use , Prevalence
SELECTION OF CITATIONS
SEARCH DETAIL
...