Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nat Immunol ; 14(8): 793-803, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23832071

ABSTRACT

The cell-to-cell transmission of viral resistance is a potential mechanism for amplifying the interferon-induced antiviral response. In this study, we report that interferon-α (IFN-α) induced the transfer of resistance to hepatitis B virus (HBV) from nonpermissive liver nonparenchymal cells (LNPCs) to permissive hepatocytes via exosomes. Exosomes from IFN-α-treated LNPCs were rich in molecules with antiviral activity. Moreover, exosomes from LNPCs were internalized by hepatocytes, which mediated the intercellular transfer of antiviral molecules. Finally, we found that exosomes also contributed to the antiviral response of IFN-α to mouse hepatitis virus A59 and adenovirus in mice. Thus, we propose an antiviral mechanism of IFN-α activity that involves the induction and intercellular transfer of antiviral molecules via exosomes.


Subject(s)
Exosomes/virology , Hepatitis B virus/immunology , Hepatitis B/immunology , Interferon-alpha/pharmacology , Liver/virology , Animals , Exosomes/immunology , Hep G2 Cells , Hepatitis B/drug therapy , Humans , Immunoblotting , Liver/immunology , Mice , Signal Transduction/immunology , Virus Replication/immunology
2.
Hepatology ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38904485

ABSTRACT

BACKGROUND AND AIMS: The surface antigen of hepatitis B virus (HBsAg) serves as an important immune-modulatory factor in chronic hepatitis B (CHB). One aspect of such modulation may act through monocytes which are the major antigen presenting cells (APCs) taking up HBsAg. There is evidence for the encapsulation of hepatocellular miRNAs by HBsAg particles, while its pathobiological significance is unclear. Here, we characterized the miRNA profile in CHB patients and probed their association with liver inflammation. APPROACHES AND RESULTS: We collected plasma from treatment-naive CHB patients (n=110) and quantified total/HBsAg-enveloped miRNAs by qRT-PCR and plasma cytokines by ELISA. The biological effects of HBsAg-delivered miRNAs in monocytes were evaluated by multiple approaches. The clinical significance of candidate miRNAs and cytokines was corroborated in patients with HBV-associated advanced liver diseases. The plasma miRNA profile showed two major clusters, one significantly associated with HBsAg titer and the other correlated with liver inflammation. Among HBsAg-carried miRNAs, miR-939 displayed most significant correlation with IL-8. Mechanistically, miR-939 in subviral particles enters monocytes and significantly augments IL-8 production via the MAPK p38 signaling pathway. Finally, the findings that miR-939 positively correlated with IL-8 level and inflammation/fibrosis stage in the cohort of HBV-associated advanced liver diseases support its causative role in the progression of liver diseases. CONCLUSION: HBsAg particles carry hepatocellular miRNAs, including miR-939, which enter monocytes and alter their functional status such as IL-8 secretion. Our findings demonstrate that HBsAg-miR-939-IL-8 axis may play a crucial role in HBV-induced hepatic necro-inflammation and progression of advanced liver diseases.

3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35131898

ABSTRACT

Type I interferons (IFN-I) exert pleiotropic biological effects during viral infections, balancing virus control versus immune-mediated pathologies, and have been successfully employed for the treatment of viral diseases. Humans express 12 IFN-alpha (α) subtypes, which activate downstream signaling cascades and result in distinct patterns of immune responses and differential antiviral responses. Inborn errors in IFN-I immunity and the presence of anti-IFN autoantibodies account for very severe courses of COVID-19; therefore, early administration of IFN-I may be protective against life-threatening disease. Here we comprehensively analyzed the antiviral activity of all IFNα subtypes against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to identify the underlying immune signatures and explore their therapeutic potential. Prophylaxis of primary human airway epithelial cells (hAEC) with different IFNα subtypes during SARS-CoV-2 infection uncovered distinct functional classes with high, intermediate, and low antiviral IFNs. In particular, IFNα5 showed superior antiviral activity against SARS-CoV-2 infection in vitro and in SARS-CoV-2-infected mice in vivo. Dose dependency studies further displayed additive effects upon coadministration with the broad antiviral drug remdesivir in cell culture. Transcriptomic analysis of IFN-treated hAEC revealed different transcriptional signatures, uncovering distinct, intersecting, and prototypical genes of individual IFNα subtypes. Global proteomic analyses systematically assessed the abundance of specific antiviral key effector molecules which are involved in IFN-I signaling pathways, negative regulation of viral processes, and immune effector processes for the potent antiviral IFNα5. Taken together, our data provide a systemic, multimodular definition of antiviral host responses mediated by defined IFN-I. This knowledge will support the development of novel therapeutic approaches against SARS-CoV-2.


Subject(s)
COVID-19 Drug Treatment , Interferon-alpha/pharmacology , SARS-CoV-2/drug effects , Transcriptome , Virus Replication/drug effects , Animals , COVID-19/immunology , COVID-19/virology , Chlorocebus aethiops , Cloning, Molecular , Disease Models, Animal , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Interferon-alpha/genetics , Interferon-alpha/immunology , Mice , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/pharmacology , Recombinant Proteins/classification , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Signal Transduction , Vero Cells
4.
Hepatology ; 75(5): 1275-1288, 2022 05.
Article in English | MEDLINE | ID: mdl-34779008

ABSTRACT

BACKGROUND AND AIMS: HBV covalently closed circular DNA (cccDNA) is a major obstacle for a cure of chronic hepatitis B. Accumulating evidence suggests that epigenetic modifications regulate the transcriptional activity of cccDNA minichromosomes. However, it remains unclear how the epigenetic state of cccDNA affects its stability. APPROACHES AND RESULTS: By using HBV infection cell models and in vitro and in vivo recombinant cccDNA (rcccDNA) and HBVcircle models, the reduction rate of HBV cccDNA and the efficacy of apolipoprotein B mRNA editing enzyme catalytic subunit 3A (APOBEC3A)-mediated and CRISPR/CRISPR-associated 9 (Cas9)-mediated cccDNA targeting were compared between cccDNAs with distinct transcriptional activities. Interferon-α treatment and hepatitis B x protein (HBx) deletion were applied as two strategies for cccDNA repression. Chromatin immunoprecipitation and micrococcal nuclease assays were performed to determine the epigenetic pattern of cccDNA. HBV cccDNA levels remained stable in nondividing hepatocytes; however, they were significantly reduced during cell division, and the reduction rate was similar between cccDNAs in transcriptionally active and transcriptionally repressed states. Strikingly, HBV rcccDNA without HBx expression exhibited a significantly longer persistence in mice. The cccDNA with low transcriptional activity exhibited an epigenetically inactive pattern and was more difficult to access by APOBEC3A and engineered CRISPR-Cas9. The epigenetic regulator activating cccDNA increased its vulnerability to APOBEC3A. CONCLUSIONS: HBV cccDNA minichromosomes in distinct epigenetic transcriptional states showed a similar reduction rate during cell division but significantly differed in their accessibility and vulnerability to targeted nucleases and antiviral agents. Epigenetic sensitization of cccDNA makes it more susceptible to damage and may potentially contribute to an HBV cure.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Animals , Cytidine Deaminase , DNA, Circular/genetics , DNA, Circular/metabolism , DNA, Viral/genetics , Epigenesis, Genetic , Hepatitis B/genetics , Hepatitis B virus/physiology , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/metabolism , Mice , Proteins , Virus Replication/genetics
5.
J Gen Virol ; 103(5)2022 05.
Article in English | MEDLINE | ID: mdl-35604380

ABSTRACT

Hepatitis B virus (HBV), which can cause chronic hepatitis B, has sophisticated machinery to establish persistent infection. Here, we report a novel mechanism whereby HBV changed miRNA packaging into extracellular vesicles (EVs) to facilitate replication. Disruption of the miRNA machinery in hepatocytes enhanced HBV replication, indicating an intrinsic miRNA-mediated antiviral state. Interference with EV release only decreased HBV replication if there was normal miRNA biogenesis, suggesting a possible link between HBV replication and EV-associated miRNAs. Microarray and qPCR analyses revealed that HBV replication changed miRNA expression in EVs. EV incubation, transfection of miRNA mimics and inhibitors, and functional pathway and network analyses showed that EV miRNAs are associated with antiviral function, suggesting that to promote survival HBV coopts EVs to excrete anti-HBV intracellular miRNAs. These data suggest a novel mechanism by which HBV maintains its replication, which has therapeutic implications.


Subject(s)
Extracellular Vesicles , MicroRNAs , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatocytes , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Virus Replication/genetics
6.
Hepatology ; 73(2): 486-502, 2021 02.
Article in English | MEDLINE | ID: mdl-32333814

ABSTRACT

BACKGROUND AND AIMS: Interferon (IFN)-α, composed of numerous subtypes, plays a crucial role in immune defense. As the most studied subtype, IFN-α2 has been used for treating chronic hepatitis B virus (HBV) infection, with advantages of finite treatment duration and sustained virologic response, but its efficacy remains relatively low. This study aimed to screen for IFN-α subtypes with the highest anti-HBV potency and to characterize mechanisms of IFN-α-mediated HBV restriction. APPROACH AND RESULTS: Using cell culture-based HBV infection systems and a human-liver chimeric mouse model, IFN-α subtype-mediated antiviral response and signaling activation were comprehensively analyzed. IFN-α14 was identified as the most effective subtype in suppression of HBV covalently closed circular DNA transcription and HBV e antigen/HBV surface antigen production, with median inhibitory concentration values approximately 100-fold lower than those of the conventional IFN-α2. IFN-α14 alone elicited IFN-α and IFN-γ signaling crosstalk in a manner similar to the combined use of IFN-α2 and IFN-γ, inducing multiple potent antiviral effectors, which synergistically restricted HBV replication. Guanylate binding protein 5, one of the most differentially expressed genes between IFN-α14-treated and IFN-α2-treated liver cells, was identified as an HBV restriction factor. A strong IFN-α-IFN-α receptor subunit 1 interaction determines the anti-HBV activity of IFN-α. The in vivo anti-HBV activity of IFN-α14 and treatment-related transcriptional patterns were further confirmed, and few adverse effects were observed. CONCLUSIONS: A concerted IFN-α and IFN-γ response in liver, which could be efficiently elicited by IFN-α subtype 14, is associated with potent HBV suppression. These data deepen the understanding of the divergent activities of IFN-α subtypes and the mechanism underlying the synergism between IFN-α and IFN-γ signaling, with implications for improved IFN therapy and HBV curative strategies.


Subject(s)
Hepatitis B virus/immunology , Hepatitis B, Chronic/drug therapy , Interferon-alpha/pharmacology , Interferon-gamma/metabolism , Animals , Disease Models, Animal , Hep G2 Cells , Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatocytes/transplantation , Humans , Interferon-alpha/genetics , Interferon-alpha/therapeutic use , Mice , Mice, Knockout , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Signal Transduction/immunology , Sustained Virologic Response , Transplantation Chimera , Virus Replication/drug effects , Virus Replication/immunology
7.
Cell Microbiol ; 22(2): e13131, 2020 02.
Article in English | MEDLINE | ID: mdl-31746509

ABSTRACT

A growing consensus indicates that host metabolism plays a vital role in viral infections. Hepatitis B virus (HBV) infection occurs in hepatocytes with active glucose metabolism and may be regulated by cellular metabolism. We addressed the question whether and how glucose regulates HBV replication in hepatocytes. The low glucose concentration at 5 mM significantly promoted HBV replication via enhanced transcription and autophagy when compared with higher glucose concentrations (10 and 25 mM). At low glucose concentration, AMPK activity was increased and led to ULK1 phosphorylation at Ser 555 and LC3-II accumulation. By contrast, the mTOR pathway was activated by high glucose concentrations, resulting in reduced HBV replication. mTOR inhibition by rapamycin reversed negative effects of high glucose concentrations on HBV replication, suggesting that low glucose concentration promotes HBV replication by stimulating the AMPK/mTOR-ULK1-autophagy axis. Consistently, we found that glucose transporters inhibition using phloretin also enhanced HBV replication via increased AMPK/mTOR-ULK1-induced autophagy. Surprisingly, the glucose analogue 2-deoxy-D-glucose reduced HBV replication through activating the Akt/mTOR signalling pathway also at the low glucose concentrations. Our study reveals that glucose is an important factor for the HBV life cycle by regulating HBV transcription and posttranscriptional steps of HBV replication via cellular autophagy.


Subject(s)
Autophagy , Glucose/metabolism , Hepatitis B virus/physiology , Hepatitis B/virology , Virus Replication , AMP-Activated Protein Kinase Kinases , Hep G2 Cells , Humans , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
8.
J Proteome Res ; 18(3): 855-864, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30550287

ABSTRACT

Exosomes are extracellular vesicles that function in intercellular communication. We have previously reported that exosomes play an important role in the transmission of antiviral molecules during interferon-α (IFN-α) treatment. In this study, the protein profiles of THP-1-derived macrophages with or without interferon-α treatment and the exosomes secreted from these cells were analyzed by label-free liquid chromatography-tandem mass spectrometry quantitation technologies. A total of 1845 and 1550 protein groups were identified in the THP-1 macrophages and the corresponding exosomes, respectively. Treating the cells with IFN-α resulted in the differential abundance of 94 proteins in cells and 67 proteins in exosomes (greater than 2.0-fold), among which 23 proteins were up-regulated in both the IFN-α treated cells and corresponding exosomes, while 14 proteins were specifically up-regulated in exosomes but not in the donor cells. GO and KEGG analysis of the identified proteins suggested that IFN-α promoted the abundance of proteins involved in the "defense response to virus" and "type I interferon signaling pathway" in both exosomes and cells. Functional analysis further indicated that exosomes from IFN-α-treated cells exhibited potent antiviral activity that restored the impaired antiviral response of IFN-α in hepatitis B virus-replicating hepatocytes. These results have deepened the understanding of the exosome-mediated transfer of IFN-α-induced antiviral molecules and may provide a new basis for therapeutic strategies to control viral infection.


Subject(s)
Exosomes/chemistry , Immunity, Innate , Interferon-alpha/pharmacology , Macrophages/metabolism , Proteomics/methods , Antiviral Agents/analysis , Antiviral Agents/metabolism , Exosomes/metabolism , Hepatitis B virus/immunology , Humans , Immunity, Innate/drug effects , Macrophages/chemistry , Macrophages/drug effects , THP-1 Cells
9.
J Virol ; 92(24)2018 12 15.
Article in English | MEDLINE | ID: mdl-30282711

ABSTRACT

Alpha interferon (IFN-α) induces the transfer of resistance to hepatitis B virus (HBV) from liver nonparenchymal cells (LNPCs) to hepatocytes via exosomes. However, little is known about the entry machinery and pathway involved in the transmission of IFN-α-induced antiviral activity. In this study, we found that macrophage exosomes uniquely depend on T cell immunoglobulin and mucin receptor 1 (TIM-1), a hepatitis A virus (HAV) receptor, to enter hepatocytes for delivering IFN-α-induced anti-HBV activity. Moreover, two primary endocytic routes for virus infection, clathrin-mediated endocytosis (CME) and macropinocytosis, collaborate to permit exosome entry and anti-HBV activity transfer. Subsequently, lysobisphosphatidic acid (LBPA), an anionic lipid closely related to endosome penetration of virus, facilitates membrane fusion of exosomes in late endosomes/multivesicular bodies (LEs/MVBs) and the accompanying exosomal cargo uncoating. Together, our findings provide comprehensive insights into the transmission route of macrophage exosomes to efficiently deliver IFN-α-induced antiviral substances and highlight the similarities between the entry mechanisms of exosomes and virus.IMPORTANCE Our previous study showed that LNPC-derived exosomes could transmit IFN-α-induced antiviral activity to HBV replicating hepatocytes, but the concrete transmission mechanisms, which include exosome entry and exosomal cargo release, remain unclear. In this study, we found that virus entry machinery and pathway were also applied to exosome-mediated cell-to-cell antiviral activity transfer. Macrophage-derived exosomes distinctively exploit hepatitis A virus receptor for access to hepatocytes. Later, CME and macropinocytosis are utilized by exosomes, followed by exosome-endosome fusion for efficient transfer of IFN-α-induced anti-HBV activity. We believe that understanding the cellular entry pathway of exosomes will be beneficial to designing exosomes as efficient vehicles for antiviral therapy.


Subject(s)
Clathrin/metabolism , Exosomes/metabolism , Hepatitis A Virus Cellular Receptor 1/metabolism , Hepatitis B virus/physiology , Interferon-alpha/metabolism , Endocytosis , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Lysophospholipids/metabolism , Macrophages/metabolism , Monoglycerides/metabolism , Pinocytosis , THP-1 Cells , Virus Internalization , Virus Replication
10.
Hepatology ; 67(4): 1237-1252, 2018 04.
Article in English | MEDLINE | ID: mdl-29059468

ABSTRACT

Interferon-α (IFN-α) is used to treat chronic hepatitis B virus (HBV) infection, but only 20%-40% of patients respond well. Clinical observations have suggested that HBV genotype is associated with the response to IFN therapy; however, its role in viral responsiveness to IFN in HBV-infected hepatocytes remains unclear. Here, we produced infectious virions of HBV genotypes A to D to infect three well-recognized cell-culture-based HBV infection systems, including primary human hepatocytes (PHH), differentiated HepaRG (dHepaRG), and HepG2-NTCP cells to quantitatively compare the antiviral effect of IFN-α on HBV across genotypes and cell models. The efficacy of IFN-α against HBV in hepatocytes was generally similar across genotypes A2, B5, C2, and D3; however, it was significantly different among the infection models given that the half maximal inhibitory concentration value of IFN-α for inhibition of viral DNA replication in PHH (<20 U/mL) and dHepaRG cells were much lower than that in HepG2-NTCP cells (>500 U/mL). Notably, even in PHH, IFN-α did not reduce HBV covalently closed circular DNA at the concentrations for which viral antigens and DNA replication intermediates were strongly reduced. The three cell-culture models exhibited differential cellular response to IFN-α. The genes reported to be associated with responsiveness to IFN-α in patients were robustly induced in PHH while weakly induced in HepG2-NTCP cells upon IFN-α treatment. Reduction or promotion of IFN response in PHH or HepG2-NTCP cells significantly attenuated or improved the inhibitory capacity of IFN-α on HBV replication, respectively. CONCLUSION: In the cell-culture-based HBV infection models, the sensitivity of HBV to IFN-α in hepatocytes is determined more by the cell-intrinsic IFN response than by viral genotype, and improvement of the IFN response in HepG2-NTCP cells promotes the efficacy of IFN-α against HBV. (Hepatology 2018;67:1237-1252).


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Hepatocytes/virology , Interferon-alpha/pharmacology , Blotting, Western , Cell Culture Techniques , DNA, Viral , Genotype , Hepatitis B/drug therapy , Hepatitis B virus/genetics , Hepatocytes/drug effects , Humans , Real-Time Polymerase Chain Reaction , Virus Replication/drug effects
11.
Mol Cell Proteomics ; 16(4 suppl 1): S144-S160, 2017 04.
Article in English | MEDLINE | ID: mdl-28242843

ABSTRACT

Hepatitis B virus (HBV) infection is a major health problem worldwide. Recent evidence suggests that some viruses can manipulate the infection process by packing specific viral and cellular components into exosomes, small nanometer-sized (30-150 nm) vesicles secreted from various cells. However, the impact of HBV replication on the content of exosomes produced by hepatocytes has not been fully delineated. In this work, an HBV-inducible cell line HepAD38 was used to directly compare changes in the protein content of exosomes secreted from HepAD38 cells with or without HBV replication. Exosomes were isolated from supernantants of HepAD38 cells cultured with or without doxycycline (dox) and their purity was confirmed by transmission electron microscopy (TEM) and Western immunoblotting assays. Ion-intensity based label-free LC-MS/MS quantitation technologies were applied to analyze protein content of exosomes from HBV replicating cells [referred as HepAD38 (dox-)-exo] and from HBV nonreplicating cells [referred as HepAD38 (dox+)-exo]. A total of 1412 exosomal protein groups were identified, among which the abundance of 35 proteins was significantly changed following HBV replication. Strikingly, 5 subunit proteins from the 26S proteasome complex, including PSMC1, PSMC2, PSMD1, PSMD7 and PSMD14 were consistently enhanced in HepAD38 (dox-)-exo. Bioinformatic analysis of differential exosomal proteins confirmed the significant enrichment of components involved in the proteasomal catabolic process. Proteasome activity assays further suggested that HepAD38 (dox-)-exo had enhanced proteolytic activity compared with HepAD38 (dox+)-exo. Furthermore, human peripheral monocytes incubated with HepAD38 (dox-)-exo induced a significantly lower level of IL-6 secretion compared with IL-6 levels from HepAD38 (dox+)-exo. Irreversible inhibition of proteasomal activity within exosomes restored higher production of IL-6 by monocytes, suggesting that transmission of proteasome subunit proteins by HepAD38 (dox-)-exo might modulate the production of pro-inflammatory molecules in the recipient monocytes. These results revealed the composition and potential function of exosomes produced during HBV replication, thus providing a new perspective on the role of exosomes in HBV-host interaction.


Subject(s)
Exosomes/virology , Hepatitis B virus/physiology , Interleukin-6/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteomics/methods , Cell Line, Tumor , Chromatography, Liquid , Exosomes/metabolism , Humans , Tandem Mass Spectrometry , Viral Proteins/metabolism , Virus Replication
12.
Article in English | MEDLINE | ID: mdl-30224536

ABSTRACT

Currently available therapies for chronic hepatitis B virus (HBV) infection can efficiently reduce viremia but induce hepatitis B surface antigen (HBsAg) loss in very few patients; also, these therapies do not greatly affect the viral covalently closed circular DNA (cccDNA). To discover new agents with complementary anti-HBV effects, we performed a drug repurposing screen of 1,018 Food and Drug Administration (FDA)-approved compounds using HBV-infected primary human hepatocytes (PHH). Several compounds belonging to the family of retinoic acid receptor (RAR) agonists were identified that reduced HBsAg levels in a dose-dependent manner without significant cytotoxicity. Among them, tazarotene exhibited the most potent anti-HBV effect, with a half-maximal inhibitory concentration (IC50) for HBsAg of less than 30 nM in PHH. The inhibitory effect was also observed in HBV-infected differentiated HepaRG (dHepaRG) models, but not in HepG2.215 cells, and HBV genotypes A to D were similarly inhibited. Tazarotene was further demonstrated to repress HBV cccDNA transcription, as determined by the levels of HBV cccDNA and RNAs and the activation of HBV promoters. Moreover, RNA sequence analysis showed that tazarotene did not induce an interferon response but altered the expression of a number of genes associated with RAR and metabolic pathways. Inhibition of RARß, but not RARα, by a specific antagonist significantly attenuated the anti-HBV activity of tazarotene, suggesting that tazarotene inhibits HBV in part through RARß. Finally, a synergistic effect of tazarotene and entecavir on HBV DNA levels was observed. Therefore, RAR agonists as represented by tazarotene were identified as potential novel anti-HBV agents.


Subject(s)
Antiviral Agents/pharmacology , Guanine/analogs & derivatives , Hepatitis B virus/drug effects , Host-Pathogen Interactions/drug effects , Nicotinic Acids/pharmacology , Receptors, Retinoic Acid/genetics , Acitretin/pharmacology , Adapalene/pharmacology , Cell Line , Cell Survival/drug effects , Dermatologic Agents/pharmacology , Drug Repositioning , Drug Synergism , Gene Expression , Guanine/pharmacology , Hep G2 Cells , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/genetics , Hepatitis B e Antigens/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/growth & development , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , High-Throughput Screening Assays , Host-Pathogen Interactions/genetics , Humans , Keratolytic Agents/pharmacology , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/metabolism , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Virus Replication/drug effects
13.
Hepatology ; 66(2): 398-415, 2017 08.
Article in English | MEDLINE | ID: mdl-28236308

ABSTRACT

Chronic hepatitis B virus (HBV) infection remains a major health problem worldwide. The covalently closed circular DNA (cccDNA) minichromosome, which serves as the template for the transcription of viral RNAs, plays a key role in viral persistence. While accumulating evidence suggests that cccDNA transcription is regulated by epigenetic machinery, particularly the acetylation of cccDNA-bound histone 3 (H3) and H4, the potential contributions of histone methylation and related host factors remain obscure. Here, by screening a series of methyltransferases and demethylases, we identified protein arginine methyltransferase 5 (PRMT5) as an effective restrictor of HBV transcription and replication. In cell culture-based models for HBV infection and in liver tissues of patients with chronic HBV infection, we found that symmetric dimethylation of arginine 3 on H4 on cccDNA was a repressive marker of cccDNA transcription and was regulated by PRMT5 depending on its methyltransferase domain. Moreover, PRMT5-triggered symmetric dimethylation of arginine 3 on H4 on the cccDNA minichromosome involved an interaction with the HBV core protein and the Brg1-based human SWI/SNF chromatin remodeler, which resulted in down-regulation of the binding of RNA polymerase II to cccDNA. In addition to the inhibitory effect on cccDNA transcription, PRMT5 inhibited HBV core particle DNA production independently of its methyltransferase activity. Further study revealed that PRMT5 interfered with pregenomic RNA encapsidation by preventing its interaction with viral polymerase protein through binding to the reverse transcriptase-ribonuclease H region of polymerase, which is crucial for the polymerase-pregenomic RNA interaction. CONCLUSION: PRMT5 restricts HBV replication through a two-part mechanism including epigenetic suppression of cccDNA transcription and interference with pregenomic RNA encapsidation; these findings improve the understanding of epigenetic regulation of HBV transcription and host-HBV interaction, thus providing new insights into targeted therapeutic intervention. (Hepatology 2017;66:398-415).


Subject(s)
DNA Replication/genetics , Hepatitis B virus/genetics , Protein-Arginine N-Methyltransferases/metabolism , Transcription, Genetic/genetics , Virus Replication/genetics , Biopsy, Needle , Blotting, Southern/methods , Cells, Cultured , DNA, Circular/genetics , DNA, Viral/genetics , Epigenomics/methods , Hep G2 Cells/cytology , Hep G2 Cells/metabolism , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/pathology , Humans , Immunohistochemistry , Immunoprecipitation , Protein-Arginine N-Methyltransferases/genetics , Real-Time Polymerase Chain Reaction/methods , Sensitivity and Specificity , Tissue Culture Techniques , Viral Load/genetics
14.
Hepatology ; 66(1): 69-83, 2017 07.
Article in English | MEDLINE | ID: mdl-28295453

ABSTRACT

Hepatitis B virus (HBV) infection shows significant gender-related differences in pathogenesis, disease progression, and development of hepatocellular carcinoma. The gender-associated differences in HBV replication and viral protein levels may be associated with distinct HBV-specific immune responses in the host. In the present study, we examined the impact of gender on HBV-specific immune responses in two different mouse models representing transient and persistent hepadnaviral infection; hydrodynamic injection with the HBV genome mimicked acute HBV infection, whereas the efficacy of therapeutic vaccination was studied in the woodchuck hepatitis virus transgenic mouse model. Consistent with previous reports, significantly higher HBV DNA and protein levels were detected in male compared to female mice. Although hydrodynamic injection with the HBV genome resulted in similar numbers of intrahepatic HBV-specific cluster of differentiation 8-positive (CD8+ ) T cells, their functionality was significantly reduced in males and correlated with higher numbers of intrahepatic regulatory T cells (Tregs). Similar effects were observed in woodchuck hepatitis virus transgenic mice immunized with a DNA prime-recombinant adenovirus boost vaccination protocol. Male mice showed functionally suppressed woodchuck hepatitis virus-specific CD8+ T-cell responses in the liver and significantly higher numbers of intrahepatic Tregs compared to females. Blockade of Treg responses in male mice led to augmented effector functions of specific CD8+ T cells and subsequently improved virus control in both models of transient and persistent hepadnaviral infection. CONCLUSION: The functionality of virus-specific CD8+ T cells in male mice was suppressed by intrahepatic Tregs and inversely correlated with levels of hepadnaviral DNA and viral protein; the induction of intrahepatic Tregs by viral replication and/or protein levels may explain the gender-related differences in the outcomes of HBV infection and limit the success of immunotherapeutic strategies in male patients. (Hepatology 2017;66:69-83).


Subject(s)
Hepatitis B virus/immunology , Hepatitis B/immunology , Immunity, Cellular/physiology , T-Lymphocytes, Regulatory/immunology , Analysis of Variance , Animals , Cells, Cultured , Disease Models, Animal , Female , Hepatitis B/prevention & control , Hepatitis B Vaccines/administration & dosage , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , Risk Factors , Sex Factors , Statistics, Nonparametric , Vaccination/methods
15.
J Virol ; 89(4): 2287-300, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25505063

ABSTRACT

UNLABELLED: The cellular innate immune system recognizing pathogen infection is essential for host defense against viruses. In parallel, viruses have developed a variety of strategies to evade the innate immunity. The hepatitis B virus (HBV), a DNA virus that causes chronic hepatitis, has been shown to inhibit RNA helicase RIG-I-mediated interferon (IFN) induction. However, it is still unknown whether HBV could affect the host DNA-sensing pathways. Here we report that in transiently HBV-transfected Huh7 cells, the stably HBV-producing cell line HepAD38, and HBV-infected HepaRG cells and primary human hepatocytes, HBV markedly interfered with IFN-ß induction and antiviral immunity mediated by the stimulator of interferon genes (STING), which has been identified as a central factor in foreign DNA recognition and antiviral innate immunity. Screening analysis demonstrated that the viral polymerase (Pol), but not other HBV-encoded proteins, was able to inhibit STING-stimulated interferon regulatory factor 3 (IRF3) activation and IFN-ß induction. Moreover, the reverse transcriptase (RT) and the RNase H (RH) domains of Pol were identified to be responsible for the inhibitory effects. Furthermore, Pol was shown to physically associate with STING and dramatically decrease the K63-linked polyubiquitination of STING via its RT domain without altering the expression level of STING. Taken together, these observations suggest that besides its inherent catalytic function, Pol has a role in suppression of IFN-ß production by direct interaction with STING and subsequent disruption of its K63-linked ubiquitination, providing a new mechanism for HBV to counteract the innate DNA-sensing pathways. IMPORTANCE: Although whether and how HBV infection induces the innate immune responses are still controversial, it has become increasingly clear that HBV has developed strategies to counteract the pattern recognition receptor-mediated signaling pathways. Previous studies have shown that type I IFN induction activated by the host RNA sensors could be inhibited by HBV. However, it remains unknown whether HBV as a DNA virus utilizes evasion mechanisms against foreign DNA-elicited antiviral signaling. In recent years, the cytosolic DNA sensor and key adaptor STING has been demonstrated to be essential in multiple foreign DNA-elicited innate immune signalings. Here, for the first time, we report STING as a new target of HBV to antagonize IFN induction and identify the viral polymerase responsible for the inhibitory effect, thus providing an additional molecular mechanism by which HBV evades the innate immunity; this implies that in addition to its inherent catalytic function, HBV polymerase is a multifunctional immunomodulatory protein.


Subject(s)
DNA, Viral/immunology , Hepatitis B virus/immunology , Hepatitis B virus/physiology , Host-Pathogen Interactions , Immune Evasion , Interferon-beta/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Cell Line , Hepatocytes/immunology , Hepatocytes/virology , Humans , Membrane Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Ubiquitination
16.
Cell Microbiol ; 17(9): 1295-303, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26243406

ABSTRACT

Viral hepatitis caused by hepatitis B virus (HBV) and hepatitis C virus (HCV) infections poses a significant burden to the public health system. Although HBV and HCV differ in structure and life cycles, they share unique characteristics, such as tropism to infect hepatocytes and association with hepatic and extrahepatic disorders that are of innate immunity nature. In response to HBV and HCV infection, the liver innate immune cells eradicate pathogens by recognizing specific molecules expressed by pathogens via distinct cellular pattern recognition receptors whose triggering activates intracellular signalling pathways inducing cytokines, interferons and anti-viral response genes that collectively function to clear infections. However, HBV and HCV evolve strategies to inactivate innate signalling factors and as such establish persistent infections without being recognized by the innate immunity. We review recent insights into how HBV and HCV are sensed and how they evade innate immunity to establish chronicity. Understanding the mechanisms of viral hepatitis is mandatory to develop effective and safe therapies for eradication of viral hepatitis.


Subject(s)
Hepacivirus/immunology , Hepatitis B virus/immunology , Hepatitis B/immunology , Hepatitis C/immunology , Host-Pathogen Interactions , Immune Evasion , Immunity, Innate , Hepatitis B/virology , Hepatitis C/virology , Humans
17.
J Immunol ; 190(10): 5142-51, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23585678

ABSTRACT

It is widely accepted that chronic hepatitis B virus (HBV) infection is the result of an ineffective antiviral immune response against HBV infection. Our previous study found that the hepatitis B surface Ag (HBsAg) was related to decreased cytokine production induced by the TLR2 ligand (Pam3csk4) in PBMCs from chronic hepatitis B patients. In this study, we further explored the mechanism involved in the inhibitory effect of HBsAg on the TLR2 signaling pathway. The results showed that both Pam3csk4-triggered IL-12p40 mRNA expression and IL-12 production in PMA-differentiated THP-1 macrophage were inhibited by HBsAg in a dose-dependent manner, but the production of IL-1ß, IL-6, IL-8, IL-10, and TNF-α was not influenced. The Pam3csk4-induced activation of NF-κB and MAPK signaling were further examined. The phosphorylation of JNK-1/2 and c-Jun was impaired in the presence of HBsAg, whereas the degradation of IκB-α, the nuclear translocation of p65, and the phosphorylation of p38 and ERK-1/2 were not affected. Moreover, the inhibition of JNK phosphorylation and IL-12 production in response to Pam3csk was observed in HBsAg-treated monocytes/macrophages (M/MΦs) from the healthy donors and the PBMCs and CD14-positive M/MΦs from chronic hepatitis B patients. Taken together, these results demonstrate that HBsAg selectively inhibits Pam3csk4- stimulated IL-12 production in M/MΦs by blocking the JNK-MAPK pathway and provide a mechanism by which HBV evades immunity and maintains its persistence.


Subject(s)
Hepatitis B Surface Antigens/metabolism , Immune Evasion/immunology , Interleukin-12/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Toll-Like Receptor 2/metabolism , Anthracenes/pharmacology , Cell Differentiation , Cell Line, Tumor , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Humans , I-kappa B Proteins , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/genetics , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , JNK Mitogen-Activated Protein Kinases/immunology , Lipopeptides/metabolism , Macrophages/metabolism , Monocytes/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptor 2/immunology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
18.
Mol Ther ; 22(2): 303-311, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24025750

ABSTRACT

The hepatitis B virus (HBV) is a DNA virus that can cause chronic hepatitis B (CHB) in humans. Current therapies for CHB infection are limited in efficacy and do not target the pre-existing viral genomic DNA, which are present in the nucleus as a covalently closed circular DNA (cccDNA) form. The transcription activator-like (TAL) effector nucleases (TALENs) are newly developed enzymes that can cleave sequence-specific DNA targets. Here, TALENs targeting the conserved regions of the viral genomic DNA among different HBV genotypes were constructed. The expression of TALENs in Huh7 cells transfected with monomeric linear full-length HBV DNA significantly reduced the viral production of HBeAg, HBsAg, HBcAg, and pgRNA, resulted in a decreased cccDNA level and misrepaired cccDNAs without apparent cytotoxic effects. The anti-HBV effect of TALENs was further demonstrated in a hydrodynamic injection-based mouse model. In addition, an enhanced antiviral effect with combinations of TALENs and interferon-α (IFN-α) treatment was observed and expression of TALENs restored HBV suppressed IFN-stimulated response element-directed transcription. Taken together, these data indicate that TALENs can specifically target and successfully inactivate the HBV genome and are potently synergistic with IFN-α, thus providing a potential therapeutic strategy for treating CHB infection.


Subject(s)
Deoxyribonucleases/metabolism , Genome, Viral , Hepatitis B virus/genetics , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Animals , Antiviral Agents/administration & dosage , Cell Line , DNA, Circular/metabolism , Deoxyribonucleases/genetics , Disease Models, Animal , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hepatitis B, Chronic/therapy , Humans , Interferon-alpha/administration & dosage , Mice , Plasmids/administration & dosage , Plasmids/genetics , Protein Binding , Viral Proteins/metabolism
19.
Hepatology ; 57(2): 470-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22996189

ABSTRACT

UNLABELLED: Treatment with exogenous interferon (IFN)-α is not effective in the majority of patients with chronic hepatitis B virus (HBV) infection. Recent evidence suggests that HBV has evolved strategies to block the nuclear translocation of signal transducer and activator of transcription (STAT) 1 to limit IFN-α-induced cellular antiviral responses. However, it remains unclear whether STAT1 translocation is impaired in chronic hepatitis B patients and what mechanisms are involved. Here we report that the expression of HBV polymerase (Pol) in human hepatic cell lines inhibited induction of IFN-stimulated genes and resulted in a weakened antiviral activity of IFN-α. Ectopic expression of Pol suppressed IFN-α-induced STAT1 serine 727 phosphorylation and STAT1/2 nuclear accumulation, whereas STAT1 tyrosine 701 phosphorylation, and STAT1-STAT2 heterodimer formation were not affected. Further studies demonstrated that Pol interacted with the catalytic domain of protein kinase C-δ (PKC-δ) and perturbed PKC-δ phosphorylation and its association with STAT1, which resulted in the suppression of STAT1 Ser727 phosphorylation. Moreover, Pol was found to interfere with nuclear transportation of STAT1/2 by competitively binding to the region of importin-α5 required for STAT1/2 recruitment. Truncation analysis suggested that the terminal protein and RNase H domains of Pol were able to bind to PKC-δ and importin-α5, respectively, and were responsible for the inhibition of IFN-α signaling. More importantly, the inhibition of STAT1 and PKC-δ phosphorylation were confirmed in a hydrodynamic-based HBV mouse model, and the blockage of IFN-α-induced STAT1/2 nuclear translocation was observed in HBV-infected cells from liver biopsies of chronic HBV patients. CONCLUSIONS: These results demonstrate a role for Pol in HBV-mediated antagonization of IFN-α signaling and provide a possible molecular mechanism by which HBV resists the IFN therapy and maintains its persistence.


Subject(s)
Interferon-alpha/therapeutic use , Protein Kinase C-delta/antagonists & inhibitors , RNA-Directed DNA Polymerase/metabolism , alpha Karyopherins/antagonists & inhibitors , Animals , Cell Line , Hep G2 Cells , Hepatitis B virus/enzymology , Humans , Interferon-alpha/pharmacology , Mice , Phosphorylation , Protein Kinase C-delta/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/antagonists & inhibitors , Vesiculovirus/drug effects
20.
Emerg Microbes Infect ; 13(1): 2287681, 2024 Dec.
Article in English | MEDLINE | ID: mdl-37994664

ABSTRACT

Type I interferons (IFN-Is) have key roles in immune defense and treatments for various diseases, including chronic hepatitis B virus (HBV) infection. All IFN-Is signal through a shared IFN-I heterodimeric receptor complex comprising IFN-α receptor 1 (IFNAR1) and IFNAR2 subunits, but differences in antiviral and immunomodulatory responses among IFN-I subtypes remain largely unknown. Because the IFN-IFNAR interactions are species-specific, mice exhibit weak responses to human IFN-I. To more fully characterize the actions of human IFN-α and its subtypes in vivo, a gene targeting strategy was employed to generate gene knock-in mice with extracellular-humanized IFNAR1/2 (IFNAR-hEC) in the C57BL/6N strain. IFNAR-hEC mice actively responded to human IFN-I, and endogenous mouse IFN-I signalling remained active in heterozygous mice (IfnarhEC/+). Analyses of IFNAR-hEC mice and isolated cells showed that human IFN-α2 and α14 subtypes exerted differential effect on the activation of JAK-STAT signalling and immune responses. Compared with IFN-α2, IFN-α14 induced greater activation of STAT1/2 and IFN-stimulated genes, synergistically elicited IFN-α and -γ signalling, and induced higher numbers of antigen-specific CD8+ T cells. Moreover, IFNAR-hEC mice with HBV replication displayed long-term viral suppression upon treatment with the clinically-used PEGylated hIFN-α2. These results indicate that IFNAR-hEC mice may be useful for elucidating antiviral and immunomodulatory functions of human IFN-Is and for conducting preclinical studies. A better understanding of the distinct activities of IFN-α subtypes can provide insights concerning the development of improved IFN-based therapy.


Subject(s)
Hepatitis B, Chronic , Interferon Type I , Humans , Mice , Animals , CD8-Positive T-Lymphocytes , Hepatitis B, Chronic/drug therapy , Mice, Inbred C57BL , Interferon-alpha , Antiviral Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL