Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
1.
Virulence ; 15(1): 2333562, 2024 12.
Article in English | MEDLINE | ID: mdl-38622757

ABSTRACT

The Picornaviridae are a large group of positive-sense, single-stranded RNA viruses, and most research has focused on the Enterovirus genus, given they present a severe health risk to humans. Other picornaviruses, such as foot-and-mouth disease virus (FMDV) and senecavirus A (SVA), affect agricultural production with high animal mortality to cause huge economic losses. The 3Dpol protein of picornaviruses is widely known to be used for genome replication; however, a growing number of studies have demonstrated its non-polymerase roles, including modulation of host cell biological processes, viral replication complex assembly and localization, autophagy, and innate immune responses. Currently, there is no effective vaccine to control picornavirus diseases widely, and clinical therapeutic strategies have limited efficiency in combating infections. Many efforts have been made to develop different types of drugs to prohibit virus survival; the most important target for drug development is the virus polymerase, a necessary element for virus replication. For picornaviruses, there are also active efforts in targeted 3Dpol drug development. This paper reviews the interaction of 3Dpol proteins with the host and the progress of drug development targeting 3Dpol.


Subject(s)
Enterovirus , Foot-and-Mouth Disease Virus , Picornaviridae Infections , Animals , Humans , Gene Products, pol/metabolism , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Virus Replication , RNA, Viral/genetics
2.
Int J Mol Sci ; 25(3)2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38339086

ABSTRACT

Acquired immunodeficiency syndrome (AIDS) is caused by human immunodeficiency virus (HIV). HIV protease, reverse transcriptase, and integrase are targets of current drugs to treat the disease. However, anti-viral drug-resistant strains have emerged quickly due to the high mutation rate of the virus, leading to the demand for the development of new drugs. One attractive target is Gag-Pol polyprotein, which plays a key role in the life cycle of HIV. Recently, we found that a combination of M50I and V151I mutations in HIV-1 integrase can suppress virus release and inhibit the initiation of Gag-Pol autoprocessing and maturation without interfering with the dimerization of Gag-Pol. Additional mutations in integrase or RNase H domain in reverse transcriptase can compensate for the defect. However, the molecular mechanism is unknown. There is no tertiary structure of the full-length HIV-1 Pol protein available for further study. Therefore, we developed a workflow to predict the tertiary structure of HIV-1 NL4.3 Pol polyprotein. The modeled structure has comparable quality compared with the recently published partial HIV-1 Pol structure (PDB ID: 7SJX). Our HIV-1 NL4.3 Pol dimer model is the first full-length Pol tertiary structure. It can provide a structural platform for studying the autoprocessing mechanism of HIV-1 Pol and for developing new potent drugs. Moreover, the workflow can be used to predict other large protein structures that cannot be resolved via conventional experimental methods.


Subject(s)
HIV Infections , HIV-1 , pol Gene Products, Human Immunodeficiency Virus , Humans , Gene Products, pol/genetics , Gene Products, pol/metabolism , HIV Infections/drug therapy , HIV Protease/genetics , HIV Protease/metabolism , HIV-1/genetics , HIV-1/metabolism , Polyproteins/genetics , RNA-Directed DNA Polymerase/metabolism , pol Gene Products, Human Immunodeficiency Virus/chemistry
3.
J Virol ; 95(19): e0044421, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34287051

ABSTRACT

DDX17 is a member of the DEAD-box helicase family proteins involved in cellular RNA folding, splicing, and translation. It has been reported that DDX17 serves as a cofactor of host zinc finger antiviral protein (ZAP)-mediated retroviral RNA degradation and exerts direct antiviral function against Raft Valley fever virus through binding to specific stem-loop structures of viral RNA. Intriguingly, we have previously shown that ZAP inhibits hepatitis B virus (HBV) replication through promoting viral RNA decay, and the ZAP-responsive element (ZRE) of HBV pregenomic RNA (pgRNA) contains a stem-loop structure, specifically epsilon, which serves as the packaging signal for pgRNA encapsidation. In this study, we demonstrated that the endogenous DDX17 is constitutively expressed in human hepatocyte-derived cells but dispensable for ZAP-mediated HBV RNA degradation. However, DDX17 was found to inhibit HBV replication primarily by reducing the level of cytoplasmic encapsidated pgRNA in a helicase-dependent manner. Immunofluorescence assay revealed that DDX17 could gain access to cytoplasm from nucleus in the presence of HBV RNA. In addition, RNA immunoprecipitation and electrophoretic mobility shift assays demonstrated that the enzymatically active DDX17 competes with HBV polymerase to bind to pgRNA at the 5' epsilon motif. In summary, our study suggests that DDX17 serves as an intrinsic host restriction factor against HBV through interfering with pgRNA encapsidation. IMPORTANCE Hepatitis B virus (HBV) chronic infection, a long-studied but yet incurable disease, remains a major public health concern worldwide. Given that HBV replication cycle highly depends on host factors, deepening our understanding of the host-virus interaction is thus of great significance in the journey of finding a cure. In eukaryotic cells, RNA helicases of the DEAD box family are highly conserved enzymes involved in diverse processes of cellular RNA metabolism. Emerging data have shown that DDX17, a typical member of the DEAD box family, functions as an antiviral factor through interacting with viral RNA. In this study, we, for the first time, demonstrate that DDX17 inhibits HBV through blocking the formation of viral replication complex, which not only broadens the antiviral spectrum of DDX17 but also provides new insight into the molecular mechanism of DDX17-mediated virus-host interaction.


Subject(s)
Capsid/metabolism , DEAD-box RNA Helicases/metabolism , Hepatitis B virus/physiology , RNA, Viral/metabolism , Virus Replication , Cell Line , Cell Line, Tumor , Cytoplasm/metabolism , DEAD-box RNA Helicases/chemistry , Gene Products, pol/metabolism , Hepatitis B virus/genetics , Humans , Nucleic Acid Conformation , Protein Domains , RNA Stability , RNA, Viral/chemistry , RNA, Viral/genetics , RNA-Binding Proteins/metabolism
4.
Viruses ; 13(3)2021 03 10.
Article in English | MEDLINE | ID: mdl-33802118

ABSTRACT

Heightened expression of human endogenous retrovirus (HERV) sequences has been associated with a range of malignancies, including prostate cancer, suggesting that they may serve as useful diagnostic or prognostic cancer biomarkers. We analysed the expression of HERV-K (Gag and Env/Np9 regions), HERV-E 4.1 (Pol and Env regions), HERV-H (Pol) and HERV-W (Gag) sequences in prostate cancer cells lines and normal prostate epithelial cells using qRT-PCR. HERV expression was also analysed in matched malignant and benign prostate tissue samples from men with prostate cancer (n = 27, median age 65.2 years (range 47-70)) and compared to prostate cancer-free male controls (n = 11). Prostate cancer epithelial cell lines exhibited a signature of HERV RNA overexpression, with all HERVs analysed, except HERV-E Pol, showing heightened expression in at least two, but more commonly all, cell lines analysed. Analysis of primary prostate material indicated increased expression of HERV-E Pol but decreased expression of HERV-E Env in both malignant and benign regions of the prostate in men with prostate cancer as compared to those without. Expression of HERV-K Gag was significantly higher in malignant regions of the prostate in men with prostate cancer as compared to matched benign regions and prostate cancer-free men (p < 0.001 for both), with 85.2% of prostate cancers donors showing malignancy-associated upregulation of HERV-K Gag RNA. HERV-K Gag protein was detected in 12/18 (66.7%) malignant tissues using immunohistochemistry, but only 1/18 (5.6%) benign tissue sections. Heightened expression of HERV-K Gag RNA and protein appears to be a sensitive and specific biomarker of prostate malignancy in this cohort of men with prostate carcinoma, supporting its potential utility as a non-invasive, adjunct clinical biomarker.


Subject(s)
Endogenous Retroviruses/genetics , Gene Products, env/genetics , Gene Products, gag/genetics , Gene Products, pol/genetics , Prostatic Neoplasms/genetics , Aged , Biomarkers, Tumor/genetics , Cell Line, Tumor , Endogenous Retroviruses/isolation & purification , Gene Expression Regulation, Neoplastic/genetics , Gene Products, env/metabolism , Gene Products, gag/metabolism , Gene Products, pol/metabolism , Humans , Male , Middle Aged , Prostate/metabolism , Prostatic Neoplasms/diagnosis
5.
J Hepatol ; 75(1): 74-85, 2021 07.
Article in English | MEDLINE | ID: mdl-33621634

ABSTRACT

BACKGROUND & AIMS: HBV remains a global threat to human health. It remains incompletely understood how HBV self-restricts in the host during most adult infections. Thus, we performed multi-omics analyses to systematically interrogate HBV-host interactions and the life cycle of HBV. METHODS: RNA-sequencing and ribosome profiling were conducted with cell-based models for HBV replication and gene expression. The novel translational events or products hereby detected were then characterized, and functionally assessed in both cell and mouse models. Moreover, quasi-species analyses of HBV subpopulations were conducted with patients at immune tolerance or activation phases, using next- or third-generation sequencing. RESULTS: We identified EnhI-SL (Enhancer I-stem loop) as a new cis element in the HBV genome; mutations disrupting EnhI-SL were found to elevate viral polymerase expression. Furthermore, while re-discovering HpZ/P', a previously under-explored isoform of HBV polymerase, we also identified HBxZ, a novel short isoform of HBX. Having confirmed their existence, we functionally characterized them as potent suppressors of HBV gene expression and genome replication. Mechanistically, HpZ/P' was found to repress HBV gene expression partially by interacting with, and sequestering SUPV3L1. Activation of the host immune system seemed to reduce the abundance of HBV mutants deficient in HpZ/P' or with disruptions in EnhI-SL. Finally, SRSF2, a host RNA spliceosome protein that is downregulated by HBV, was found to promote the splicing of viral pre-genomic RNA and HpZ/P' biogenesis. CONCLUSION: This study has identified multiple self-restricting HBV-host interactions. In particular, SRSF2-HpZ/P' appeared to constitute another negative feedback mechanism in the HBV life cycle. Targeting host splicing machinery might thus represent a strategy to intervene in HBV-host interactions. LAY SUMMARY: There remain many unknowns about the natural history of HBV infection in adults. Herein, we identified new HBV-host mechanisms which could be responsible for self-restricting infections. Targeting these mechanisms could be a promising strategy for the treatment of HBV infections.


Subject(s)
Gene Products, pol/metabolism , Hepatitis B virus , Hepatitis B, Chronic , Host Microbial Interactions/immunology , Virus Replication , Animals , Drug Discovery , Genome, Viral/physiology , Hepatitis B virus/enzymology , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Humans , Mice , Promoter Regions, Genetic , Protein Modification, Translational , RNA, Ribosomal, Self-Splicing/metabolism , RNA-Directed DNA Polymerase/metabolism , Serine-Arginine Splicing Factors/metabolism , Virus Replication/genetics , Virus Replication/immunology
6.
Genes Cells ; 25(8): 523-537, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32415897

ABSTRACT

Although several nucleo(s)tide analogs are available for treatment of HBV infection, long-term treatment with these drugs can lead to the emergence of drug-resistant viruses. Recent HIV-1 studies suggest that combination therapies using nucleo(s)tide reverse transcriptase inhibitors (NRTIs) and non-nucleo(s)tide reverse transcriptase inhibitors (NNRTIs) could drastically inhibit the viral genome replication of NRTI-resistant viruses. In order to carry out such combinational therapy against HBV, several new NRTIs and NNRTIs should be developed. Here, we aimed to identify novel NNRTIs targeting the HBV polymerase terminal protein (TP)-reverse transcriptase (RT) (TP-RT) domain, which is a critical domain for HBV replication. We expressed and purified the HBV TP-RT with high purity using an Escherichia coli expression system and established an in vitro ε RNA-binding assay system. Then, we used TP-RT in cell-free assays to screen candidate inhibitors from a chemical compound library, and identified two compounds, 6-hydroxy-DL-DOPA and N-oleoyldopamine, which inhibited the binding of ε RNA with the HBV polymerase. Furthermore, these drugs reduced HBV DNA levels in cell-based assays as well by inhibiting packaging of pregenome RNA into capsids. The novel screening system developed herein should open a new pathway the discovery of drugs targeting the HBV TP-RT domain to treat HBV infection.


Subject(s)
Drug Evaluation, Preclinical/methods , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication/drug effects , Carrier Proteins/metabolism , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , Gene Products, pol/genetics , Gene Products, pol/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Humans , Protein Binding , RNA/metabolism , RNA-Binding Motifs/genetics , RNA-Directed DNA Polymerase/chemistry , RNA-Directed DNA Polymerase/genetics , Small Molecule Libraries
7.
Sci Rep ; 10(1): 263, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937823

ABSTRACT

Koala retrovirus (KoRV) displays features of both an endogenous and exogenous virus and is linked to neoplasia and immunosuppression in koalas. This study explores the apparent differences in the nature and impact of KoRV infection between geographically and genetically separated "northern" and "southern" koala populations, by investigating the disease status, completeness of the KoRV genome and the proviral (DNA) and viral (RNA) loads of 71 northern and 97 southern koalas. All northern animals were positive for all KoRV genes (gag, pro-pol and env) in both DNA and RNA forms, whereas many southern animals were missing one or more KoRV genes. There was a significant relationship between the completeness of the KoRV genome and clinical status in this population. The proviral and viral loads of the northern population were significantly higher than those of the southern population (P < 0.0001), and many provirus-positive southern animals failed to express any detectable KoRV RNA. Across both populations there was a positive association between proviral load and neoplasia (P = 0.009). Potential reasons for the differences in the nature of KoRV infection between the two populations are discussed.


Subject(s)
Retroviridae Infections/pathology , Retroviridae/genetics , Aging/genetics , Animals , Australia/epidemiology , DNA/metabolism , Female , Gene Products, env/genetics , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Gene Products, pol/genetics , Gene Products, pol/metabolism , Male , Phascolarctidae , Proviruses/genetics , RNA, Viral/blood , Retroviridae/isolation & purification , Retroviridae Infections/epidemiology , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Viral Load
8.
Am J Physiol Gastrointest Liver Physiol ; 318(1): G162-G173, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31604033

ABSTRACT

Hepatitis B virus (HBV) exploits multiple strategies to evade host immune surveillance. Programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) signaling plays a critical role in regulating T cell homeostasis. However, it remains largely unknown as to how HBV infection elevates PD-L1 expression in hepatocytes. A mouse model of HBV infection was established by hydrodynamic injection with a vector containing 1.3-fold overlength HBV genome (pHBV1.3) via the tail vein. Coculture experiments with HBV-expressing hepatoma cells and Jurkat T cells were established in vitro. We observed significant decrease in the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and increase in ß-catenin/PD-L1 expression in liver tissues from patients with chronic hepatitis B and mice subjected to pHBV1.3 hydrodynamic injection. Mechanistically, decrease in PTEN enhanced ß-catenin/c-Myc signaling and PD-L1 expression in HBV-expressing hepatoma cells, which in turn augmented PD-1 expression, lowered IL-2 secretion, and induced T cell apoptosis. However, ß-catenin disruption inhibited PTEN-mediated PD-L1 expression, which was accompanied by decreased PD-1 expression, and increased IL-2 production in T cells. Luciferase reporter assays revealed that c-Myc stimulated transcriptional activity of PD-L1. In addition, HBV X protein (HBx) and HBV polymerase (HBp) contributed to PTEN downregulation and ß-catenin/PD-L1 upregulation. Strikingly, PTEN overexpression in hepatocytes inhibited ß-catenin/PD-L1 signaling and promoted HBV clearance in vivo. Our findings suggest that HBV-triggered PTEN/ß-catenin/c-Myc signaling via HBx and HBp enhances PD-L1 expression, leading to inhibition of T cell response, and promotes HBV immune evasion.NEW & NOTEWORTHY This study demonstrates that during HBV infection, HBV can increase PD-L1 expression via PTEN/ß-catenin/c-Myc signaling pathway, which in turn inhibits T cell response and ultimately promotes HBV immune evasion. Targeting this signaling pathway is a potential strategy for immunotherapy of chronic hepatitis B.


Subject(s)
B7-H1 Antigen/metabolism , Hepatitis B virus/metabolism , Hepatitis B, Chronic/metabolism , Hepatocytes/enzymology , Immune Evasion , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-myc/metabolism , T-Lymphocytes/enzymology , beta Catenin/metabolism , Animals , Coculture Techniques , Disease Models, Animal , Gene Products, pol/genetics , Gene Products, pol/metabolism , Hep G2 Cells , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatocytes/immunology , Hepatocytes/virology , Humans , Jurkat Cells , Lymphocyte Activation , Male , Mice, Inbred BALB C , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/virology , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins
9.
Cell ; 179(3): 632-643.e12, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31607510

ABSTRACT

Antisense Piwi-interacting RNAs (piRNAs) guide silencing of established transposons during germline development, and sense piRNAs drive ping-pong amplification of the antisense pool, but how the germline responds to genome invasion is not understood. The KoRV-A gammaretrovirus infects the soma and germline and is sweeping through wild koalas by a combination of horizontal and vertical transfer, allowing direct analysis of retroviral invasion of the germline genome. Gammaretroviruses produce spliced Env mRNAs and unspliced transcripts encoding Gag, Pol, and the viral genome, but KoRV-A piRNAs are almost exclusively derived from unspliced genomic transcripts and are strongly sense-strand biased. Significantly, selective piRNA processing of unspliced proviral transcripts is conserved from insects to placental mammals. We speculate that bypassed splicing generates a conserved molecular pattern that directs proviral genomic transcripts to the piRNA biogenesis machinery and that this "innate" piRNA response suppresses transposition until antisense piRNAs are produced, establishing sequence-specific adaptive immunity.


Subject(s)
Gammaretrovirus/genetics , Phascolarctidae/genetics , RNA, Small Interfering/genetics , Animals , DNA Transposable Elements , Gammaretrovirus/metabolism , Gammaretrovirus/pathogenicity , Gene Products, env/genetics , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Gene Products, pol/genetics , Gene Products, pol/metabolism , Genome , Germ Cells/metabolism , Germ Cells/virology , Male , Mice , Mice, Inbred C57BL , Phascolarctidae/virology , RNA Splicing , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Small Interfering/metabolism
10.
Liver Int ; 39(12): 2273-2284, 2019 12.
Article in English | MEDLINE | ID: mdl-31419377

ABSTRACT

BACKGROUND & AIMS: Hepatitis B virus (HBV) infection is the most critical factor underlying liver cirrhosis and hepatocellular carcinoma worldwide. IL-1ß and IL-18, generated by activation of the inflammasome/caspase-1 signaling pathway, play important roles in the control and clearance of HBV. However, the specific relationship between the inflammasome response and IFN-α resistance or viral persistence is yet to be established. METHODS: Blood samples of patients and supernatant fractions of HBV cell lines were collected for analysis and the effects on inflammasome activation and IL-1ß production evaluated via enzyme-linked immunosorbent assay (ELISA), western blot, quantitative RT-PCR and immunofluorescence. RESULTS: IL-1ß and IL-18 levels produced in sera of IFN-α non-responders were significantly lower than those of responders and normal donors. Additionally, expression of IL-1ß and inflammasome components was decreased in peripheral blood mononuclear cells (PBMC) of non-responders, compared with those of responders. In vitro experiments on HepG2, HepG2.2.15 and HepAD38 cell lines showed that HBV induces a significant decrease in IL-1ß production through inhibiting activation of the NF-κB signaling and inflammasome/caspase-1 pathways. And hepatitis B virus polymerase (HBV-Pol) appeared crucial for these inhibitory effects of HBV. CONCLUSION: IL-1ß production is suppressed in HBV carriers and IFN-α non-responders. HBV induces a significant decrease in IL-1ß production through inhibiting the NF-κB signaling and inflammasome pathways, for which HBV-Pol is a crucial requirement. Trial approval number: 20 173 402.


Subject(s)
Gene Products, pol/metabolism , Hepatitis B, Chronic/blood , Host-Pathogen Interactions , Inflammasomes/metabolism , Interleukin-1beta/blood , Adult , Antiviral Agents/therapeutic use , Case-Control Studies , Female , Hep G2 Cells , Hepatitis B, Chronic/drug therapy , Hepatitis C, Chronic/blood , Humans , Interferon-alpha/therapeutic use , Interleukin-18/blood , Male , Middle Aged
11.
Sci Adv ; 5(2): eaau7130, 2019 02.
Article in English | MEDLINE | ID: mdl-30775435

ABSTRACT

About 257 million people with chronic infection of hepatitis B virus (HBV) worldwide are at high risk of developing terminal liver diseases. Reactivation of virus replication has been frequently reported in those patient populations receiving imatinib (an Abl kinase inhibitor) or bortezomib (a proteasome inhibitor) to treat concurrent diseases, but the underlying mechanism for this reactivation is unknown. We report that the HBV polymerase protein is recruited by Cdt2 to the cullin-RING ligase 4 (CRL4) for ubiquitination and proteasome degradation and that this process is stimulated by the c-Abl nonreceptor tyrosine kinase. Genetic ablation of the Abl-CRL4Cdt2 axis or pharmaceutical inhibition of this process stabilizes HBV polymerase protein and increases viral loads in HBV-infected liver cancer cell lines. Our study reveals a kinase-dependent activation of CRL4 ubiquitin ligase that can be targeted for blocking HBV replication.


Subject(s)
Gene Products, pol/metabolism , Hepatitis B virus/physiology , Hepatitis B/metabolism , Hepatitis B/virology , Host-Pathogen Interactions , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Virus Replication , Cell Line, Tumor , Enzyme Stability , Humans , Models, Biological , Nuclear Proteins/metabolism , Protein Binding , Proteolysis , Substrate Specificity , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Gut ; 68(5): 905-915, 2019 05.
Article in English | MEDLINE | ID: mdl-30622109

ABSTRACT

OBJECTIVE: A hallmark of chronic HBV (cHBV) infection is the presence of impaired HBV-specific CD8+ T cell responses. Functional T cell exhaustion induced by persistent antigen stimulation is considered a major mechanism underlying this impairment. However, due to their low frequencies in chronic infection, it is currently unknown whether HBV-specific CD8+ T cells targeting different epitopes are similarly impaired and share molecular profiles indicative of T cell exhaustion. DESIGN: By applying peptide-loaded MHC I tetramer-based enrichment, we could detect HBV-specific CD8+ T cells targeting epitopes in the HBV core and the polymerase proteins in the majority of 85 tested cHBV patients with low viral loads. Lower detection rates were obtained for envelope-specific CD8+ T cells. Subsequently, we performed phenotypic and functional in-depth analyses. RESULTS: HBV-specific CD8+ T cells are not terminally exhausted but rather exhibit a memory-like phenotype in patients with low viral load possibly reflecting weak ongoing cognate antigen recognition. Moreover, HBV-specific CD8+ T cells targeting core versus polymerase epitopes significantly differed in frequency, phenotype and function. In particular, in comparison with core-specific CD8+ T cells, a higher frequency of polymerase-specific CD8+ T cells expressed CD38, KLRG1 and Eomes accompanied by low T-bet expression and downregulated CD127 indicative of a more severe T cell exhaustion. In addition, polymerase-specific CD8+ T cells exhibited a reduced expansion capacity that was linked to a dysbalanced TCF1/BCL2 expression. CONCLUSIONS: Overall, the molecular mechanisms underlying impaired T cell responses differ with respect to the targeted HBV antigens. These results have potential implications for immunotherapeutic approaches in HBV cure.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Gene Products, pol/metabolism , Hepatitis B virus/immunology , Hepatitis B, Chronic/metabolism , Viral Core Proteins/metabolism , Viral Load , Adult , Aged , Cohort Studies , Female , Hepatitis B, Chronic/etiology , Humans , Male , Middle Aged , Phenotype
13.
PLoS One ; 13(5): e0197664, 2018.
Article in English | MEDLINE | ID: mdl-29782545

ABSTRACT

Current therapeutics for hepatitis B virus (HBV) patients such as nucleoside analogs (NAs) are effective; however, new antiviral drugs against HBV are still desired. Since the interaction between the epsilon (ε) sequence of HBV pregenomic RNA and viral polymerase (Pol) is a key step in the HBV replication cycle, we aimed to identify small compounds for its inhibition, and established a pull-down assay system for the detection of ε-RNA-binding-Pol. Screening showed that 5 out of 3,965 compounds inhibited ε-Pol binding, and we identified rosmarinic acid, which exhibited specificity, as a potential antiviral agent. In order to examine the anti-HBV effects of rosmarinic acid, HBV-infected primary human hepatocytes from a humanized mouse liver were treated with rosmarinic acid. The rosmarinic acid treatment decreased HBV components including the amounts of extracellular HBV DNA with negligible cytotoxicity. We also investigated the combined effects of rosmarinic acid and the NA, lamivudine. rosmarinic acid slightly enhanced the anti-HBV activity of lamivudine, suggesting that the HBV replication step targeted by rosmarinic acid is distinct from that of NA. We analyzed an additional 25 rosmarinic acid derivatives, and found that 5 also inhibited ε-Pol. Structural comparisons between these derivatives implied that the "two phenolic hydroxyl groups at both ends" and the "caffeic acid-like structure" of rosmarinic acid are critical for the inhibition of ε-Pol binding. Collectively, our results demonstrate that rosmarinic acid inhibits HBV replication in HBV-infected cells by specifically targeting ε-Pol binding.


Subject(s)
Antiviral Agents/pharmacology , Cinnamates/pharmacology , Depsides/pharmacology , Hepatitis B virus/drug effects , Virus Replication/drug effects , Animals , Antiviral Agents/administration & dosage , Cells, Cultured , Cinnamates/administration & dosage , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Depsides/administration & dosage , Drug Synergism , Enzyme Inhibitors/pharmacology , Gene Products, pol/antagonists & inhibitors , Gene Products, pol/metabolism , HEK293 Cells , Hep G2 Cells , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Lamivudine/administration & dosage , Mice , Quercetin/pharmacology , RNA, Viral/genetics , RNA, Viral/metabolism , Rosmarinic Acid
14.
Bioorg Med Chem ; 26(8): 1573-1578, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29459146

ABSTRACT

In this study, a series of 2-arylthio-5-iodo pyrimidine derivatives, as non-nucleoside hepatitis B virus inhibitors, were evaluated and firstly reported as potential anti-HBV agents. To probe the mechanism of active agents, DHBV polymerase was isolated and a non-radioisotopic assay was established for measuring HBV polymerase. The biological results demonstrated that 2-arylthio-5-iodo pyrimidine derivatives targeted HBV polymerase. In addition, pharmacophore models were constructed for future optimization of lead compounds. Further study will be performed for the development of non-nucleoside anti-HBV agents.


Subject(s)
Antiviral Agents/pharmacology , Gene Products, pol/antagonists & inhibitors , Hepatitis B virus/drug effects , Pyrimidines/pharmacology , Sulfhydryl Compounds/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Ducks , Gene Products, pol/metabolism , Hep G2 Cells , Humans , Liver/virology , Microbial Sensitivity Tests , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors , Structure-Activity Relationship , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry
15.
J Biol Chem ; 292(48): 19814-19825, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28972144

ABSTRACT

The pyridine-based multimerization selective HIV-1 integrase (IN) inhibitors (MINIs) are a distinct subclass of allosteric IN inhibitors. MINIs potently inhibit HIV-1 replication during virion maturation by inducing hyper- or aberrant IN multimerization but are largely ineffective during the early steps of viral replication. Here, we investigated the mechanism for the evolution of a triple IN substitution (T124N/V165I/T174I) that emerges in cell culture with a representative MINI, KF116. We show that HIV-1 NL4-3(IN T124N/V165I/T174I) confers marked (>2000-fold) resistance to KF116. Two IN substitutions (T124N/T174I) directly weaken inhibitor binding at the dimer interface of the catalytic core domain but at the same time markedly impair HIV-1 replication capacity. Unexpectedly, T124N/T174I IN substitutions inhibited proteolytic processing of HIV-1 polyproteins Gag and Gag-Pol, resulting in immature virions. Strikingly, the addition of the third IN substitution (V165I) restored polyprotein processing, virus particle maturation, and significant levels of replication capacity. These results reveal an unanticipated role of IN for polyprotein proteolytic processing during virion morphogenesis. The complex evolutionary pathway for the emergence of resistant viruses, which includes the need for the compensatory V165I IN substitution, highlights a relatively high genetic barrier exerted by MINI KF116. Additionally, we have solved the X-ray structure of the drug-resistant catalytic core domain protein, which provides means for rational development of second-generation MINIs.


Subject(s)
Gene Products, gag/metabolism , Gene Products, pol/metabolism , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Pyridines/chemistry , Crystallography, X-Ray , Drug Resistance, Viral , HEK293 Cells , HIV Integrase Inhibitors/chemistry , HIV-1/metabolism , Humans , Proteolysis , Surface Plasmon Resonance
16.
PLoS One ; 12(7): e0181012, 2017.
Article in English | MEDLINE | ID: mdl-28700677

ABSTRACT

Adenovirus (Ad) vectors deleted of the early region 1 (E1) are widely used for transgene delivery in preclinical and clinical gene therapy studies. Although proteins encoded within the E1 region are required for efficient virus replication, previous studies have suggested that certain viral or cellular proteins can functionally compensate for E1, leading to expression of the early region 2 (E2)-encoded replicative proteins and subsequent virus replication. We have generated a series of E1-encoding and E1-deficient Ad vectors containing a FLAG-epitope tag on each of the E2-encoded proteins: DNA-binding protein (DBP), terminal protein (TP) and DNA polymerase (Pol). Using these constructs, we show that for the replication-competent virus, the expression level of each E2-encoded protein declines with increasing distance from the E2 promoter, with E2A-encoded DBP expression being ~800-fold higher than E2B-encoded TP. Pol was expressed at extremely low levels in infected cells, and immunoprecipitation from cell lysates was required prior to its detection by immunoblot. We further show that DBP was expressed 200- to 400-fold less efficiently from an E1-deficient virus compared to a replication-competent virus in A549 and HepG2 cells, which was accompanied by a very small increase in genome copy number. For the E1-deficient virus, late gene expression (a marker of virus replication) was only observed at very high multiplicities of infection. These data show that E1-deleted Ad gives rise to limited expression of the E2-encoded genes and replication in infected cells, but highlight the importance of considering viral dose-dependent effects in gene therapy studies.


Subject(s)
Adenoviruses, Human/genetics , DNA Replication/physiology , Genetic Vectors/genetics , Adenoviruses, Human/physiology , Cell Line , DNA Replication/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Products, pol/genetics , Gene Products, pol/metabolism , Hep G2 Cells , Humans , Viral Proteins/genetics , Viral Proteins/metabolism
17.
Virology ; 506: 28-33, 2017 06.
Article in English | MEDLINE | ID: mdl-28314126

ABSTRACT

Foamy viruses are non-pathogenic retroviruses and represent a tool for vector development. For gene therapy applications and for analyses of viral protein composition infectious particles need to be purified, which has been difficult for foamy viruses in the past. Here, we describe a novel, simple, and fast purification method for prototype foamy viruses with high purity using size exclusion and affinity chromatography. More than 99,9% of the contaminating proteins were removed. The purified viruses were used to determine the amount of the incorporated Pol protein relative to Gag. The determined Gag to Pol PR-RT ratio of 30:1 confirmed previous studies suggesting FV virions encapsidate fewer number of Pol molecules than orthoretroviruses.


Subject(s)
Chromatography, Affinity/methods , Chromatography, Gel/methods , Spumavirus/growth & development , Virion/growth & development , Gene Expression Regulation, Viral , Gene Products, pol/genetics , Gene Products, pol/metabolism , Humans , Retroviridae Infections/virology , Spumavirus/genetics , Spumavirus/isolation & purification , Spumavirus/physiology , Virion/genetics , Virion/isolation & purification , Virion/physiology , Virus Assembly , Virus Cultivation
18.
Methods Mol Biol ; 1540: 157-177, 2017.
Article in English | MEDLINE | ID: mdl-27975315

ABSTRACT

The hepatitis B virus (HBV) polymerase synthesizes the viral DNA genome from the pre-genomic RNA (pgRNA) template through reverse transcription. Initiation of viral DNA synthesis is accomplished via a novel protein priming mechanism, so named because the polymerase itself acts as a primer, whereby the initiating nucleotide becomes covalently linked to a tyrosine residue on the viral polymerase. Protein priming, in turn, depends on specific recognition of the packaging signal on pgRNA called epsilon. These early events in viral DNA synthesis can now be dissected in vitro as described here.The polymerase is expressed in mammalian cells and purified by immunoprecipitation. The purified protein is associated with host cell factors, is enzymatically active, and its priming activity is epsilon dependent. A minimal epsilon RNA construct from pgRNA is co-expressed with the polymerase in cells. This RNA binds to and co-immunoprecipitates with the polymerase. Modifications can be made to either the epsilon RNA or the polymerase protein by manipulating the expression plasmids. Also, the priming reaction itself can be modified to assay for the initiation or subsequent DNA synthesis during protein priming, the susceptibility of the polymerase to chemical inhibitors, and the precise identification of the DNA products upon their release from the polymerase. The identity of associated host factors can also be evaluated. This protocol closely mirrors our current understanding of the RNA binding and protein priming steps of the HBV replication cycle, and it is amenable to modification. It should therefore facilitate both basic research and drug discovery.


Subject(s)
Gene Products, pol/metabolism , Hepatitis B virus/physiology , RNA, Viral/genetics , Transcription, Genetic , Cell Line , DNA Cleavage , DNA-Binding Proteins , Enzyme Activation , Gene Expression , Gene Products, pol/genetics , Gene Products, pol/isolation & purification , Hepatitis B virus/enzymology , Humans , In Vitro Techniques , Multiprotein Complexes , Nuclear Proteins/metabolism , Phosphoric Diester Hydrolases , Plasmids/genetics , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reverse Transcription , Transcription Factors/metabolism , Transfection , Virus Replication
19.
J Virol ; 91(3)2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27852858

ABSTRACT

Hepatitis B virus (HBV) encodes a multifunction reverse transcriptase or polymerase (P), which is composed of several domains. The terminal protein (TP) domain is unique to HBV and related hepadnaviruses and is required for specifically binding to the viral pregenomic RNA (pgRNA). Subsequently, the TP domain is necessary for pgRNA packaging into viral nucleocapsids and the initiation of viral reverse transcription for conversion of the pgRNA to viral DNA. Uniquely, the HBV P protein initiates reverse transcription via a protein priming mechanism using the TP domain as a primer. No structural homologs or high-resolution structure exists for the TP domain. Secondary structure prediction identified three disordered loops in TP with highly conserved sequences. A meta-analysis of mutagenesis studies indicated these predicted loops are almost exclusively where functionally important residues are located. Newly constructed TP mutations revealed a priming loop in TP which plays a specific role in protein-primed DNA synthesis beyond simply harboring the site of priming. Substitutions of potential sites of phosphorylation surrounding the priming site demonstrated that these residues are involved in interactions critical for priming but are unlikely to be phosphorylated during viral replication. Furthermore, the first 13 and 66 TP residues were shown to be dispensable for protein priming and pgRNA binding, respectively. Combining current and previous mutagenesis work with sequence analysis has increased our understanding of TP structure and functions by mapping specific functions to distinct predicted secondary structures and will facilitate antiviral targeting of this unique domain. IMPORTANCE: HBV is a major cause of viral hepatitis, liver cirrhosis, and hepatocellular carcinoma. One important feature of this virus is its polymerase, the enzyme used to create the DNA genome from a specific viral RNA by reverse transcription. One region of this polymerase, the TP domain, is required for association with the viral RNA and production of the DNA genome. Targeting the TP domain for antiviral development is difficult due to the lack of homology to other proteins and high-resolution structure. This study mapped the TP functions according to predicted secondary structure, where it folds into alpha helices or unstructured loops. Three predicted loops were found to be the most important regions functionally and the most conserved evolutionarily. Identification of these functional subdomains in TP will facilitate its targeting for antiviral development.


Subject(s)
Gene Products, pol/genetics , Gene Products, pol/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Protein Interaction Domains and Motifs , Amino Acid Sequence , Conserved Sequence , Gene Products, pol/chemistry , Humans , Models, Molecular , Mutation , Phenotype , Protein Conformation, alpha-Helical , RNA, Messenger/genetics , RNA, Viral , RNA-Binding Proteins
20.
Viruses ; 8(7)2016 07 07.
Article in English | MEDLINE | ID: mdl-27399760

ABSTRACT

Nef proteins from all primate Lentiviruses, including the simian immunodeficiency virus of chimpanzees (SIVcpz), increase viral progeny infectivity. However, the function of Nef involved with the increase in viral infectivity is still not completely understood. Nonetheless, until now, studies investigating the functions of Nef from SIVcpz have been conducted in the context of the HIV-1 proviruses. In an attempt to investigate the role played by Nef during the replication cycle of an SIVcpz, a Nef-defective derivative was obtained from the SIVcpzWTGab2 clone by introducing a frame shift mutation at a unique restriction site within the nef sequence. This nef-deleted clone expresses an N-terminal 74-amino acid truncated peptide of Nef and was named SIVcpz-tNef. We found that the SIVcpz-tNef does not behave as a classic nef-deleted HIV-1 or simian immunodeficiency virus of macaques SIVmac. Markedly, SIVcpz-tNef progeny from both Hek-293T and Molt producer cells were completely non-infectious. Moreover, the loss in infectivity of SIVcpz-tNef correlated with the inhibition of Gag and GagPol processing. A marked accumulation of Gag and very low levels of reverse transcriptase were detected in viral lysates. Furthermore, these observations were reproduced once the tNef peptide was expressed in trans both in SIVcpzΔNef and HIV-1WT expressing cells, demonstrating that the truncated peptide is a dominant negative for viral processing and infectivity for both SIVcpz and HIV-1. We demonstrated that the truncated Nef peptide binds to GagPol outside the protease region and by doing so probably blocks processing of both GagPol and Gag precursors at a very early stage. This study demonstrates for the first time that naturally-occurring Nef peptides can potently block lentiviral processing and infectivity.


Subject(s)
Gene Products, nef/metabolism , HIV-1/physiology , Simian Immunodeficiency Virus/physiology , Virus Replication , Animals , Cell Line , Frameshift Mutation , Gene Knockout Techniques , Gene Products, gag/metabolism , Gene Products, nef/genetics , Gene Products, pol/metabolism , Humans , Pan troglodytes , Protein Binding , Simian Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...