Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Virol ; 97(10): e0072223, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37754761

ABSTRACT

IMPORTANCE: Chronic hepatitis B is the most important cause of liver cancer worldwide and affects more than 290 million people. Current treatments are mostly suppressive and rarely lead to a cure. Therefore, there is a need for novel and curative drugs that target the host or the causative agent, hepatitis B virus itself. Capsid assembly modulators are an interesting class of antiviral molecules that may one day become part of curative treatment regimens for chronic hepatitis B. Here we explore the characteristics of a particularly interesting subclass of capsid assembly modulators. These so-called non-HAP CAM-As have intriguing properties in cell culture but also clear virus-infected cells from the mouse liver in a gradual and sustained way. We believe they represent a considerable improvement over previously reported molecules and may one day be part of curative treatment combinations for chronic hepatitis B.


Subject(s)
Antiviral Agents , Capsid , Hepatitis B virus , Hepatitis B, Chronic , Virus Assembly , Animals , Humans , Mice , Antiviral Agents/classification , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Capsid/chemistry , Capsid/drug effects , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Cells, Cultured , Hepatitis B virus/chemistry , Hepatitis B virus/drug effects , Hepatitis B virus/growth & development , Hepatitis B virus/metabolism , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , In Vitro Techniques , Virus Assembly/drug effects , Disease Models, Animal
2.
PLoS Pathog ; 18(1): e1010204, 2022 01.
Article in English | MEDLINE | ID: mdl-35030230

ABSTRACT

The hepatitis B virus (HBV) core protein (HBc) functions in multiple steps of the viral life cycle. Heteroaryldihydropyrimidine compounds (HAPs) such as Bay41-4109 are capsid protein allosteric modulators that accelerate HBc degradation and inhibit the virion secretion of HBV, specifically by misleading HBc assembly into aberrant non-capsid polymers. However, the subsequent cellular fates of these HAP-induced aberrant non-capsid polymers are not well understood. Here, we discovered that that the chaperone-binding E3 ubiquitin ligase protein STUB1 is required for the removal of Bay41-4109-induced aberrant non-capsid polymers from HepAD38 cells. Specifically, STUB1 recruits BAG3 to transport Bay41-4109-induced aberrant non-capsid polymers to the perinuclear region of cells, thereby initiating p62-mediated macroautophagy and lysosomal degradation. We also demonstrate that elevating the STUB1 level enhances the inhibitory effect of Bay41-4109 on the production of HBeAg and HBV virions in HepAD38 cells, in HBV-infected HepG2-NTCP cells, and in HBV transgenic mice. STUB1 overexpression also facilitates the inhibition of Bay41-4109 on the cccDNA formation in de novo infection of HBV. Understanding these molecular details paves the way for applying HAPs as a potentially curative regimen (or a component of a combination treatment) for eradicating HBV from hepatocytes of chronic infection patients.


Subject(s)
Antiviral Agents/pharmacology , Capsid Proteins/drug effects , Hepatitis B virus/drug effects , Hepatitis B virus/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Capsid Proteins/metabolism , Hepatitis B/virology , Humans , Macroautophagy/drug effects , Mice
3.
Molecules ; 26(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34641294

ABSTRACT

Supramolecular protein complexes are the corner stone of biological processes; they are essential for many biological functions. Unraveling the interactions responsible for the (dis)assembly of these complexes is required to understand nature and to exploit such systems in future applications. Virus capsids are well-defined assemblies of hundreds of proteins and form the outer shell of non-enveloped viruses. Due to their potential as a drug carriers or nano-reactors and the need for virus inactivation strategies, assessing the intactness of virus capsids is of great interest. Current methods to evaluate the (dis)assembly of these protein assemblies are experimentally demanding in terms of instrumentation, expertise and time. Here we investigate a new strategy to monitor the disassembly of fluorescently labeled virus capsids. To monitor surfactant-induced capsid disassembly, we exploit the complex photophysical interplay between multiple fluorophores conjugated to capsid proteins. The disassembly of the capsid changes the photophysical interactions between the fluorophores, and this can be spectrally monitored. The presented data show that this low complexity method can be used to study and monitor the disassembly of supramolecular protein complexes like virus capsids. However, the range of labeling densities that is suitable for this assay is surprisingly narrow.


Subject(s)
Capsid/chemistry , Fluorescent Dyes/chemistry , Surface-Active Agents/adverse effects , Capsid/drug effects , Capsid Proteins/chemistry , Capsid Proteins/drug effects , Fluorescence Resonance Energy Transfer , Protein Conformation , Virus Inactivation
4.
PLoS Pathog ; 17(4): e1009434, 2021 04.
Article in English | MEDLINE | ID: mdl-33878123

ABSTRACT

The glycosphingolipid (GSL) globoside (Gb4) is essential for parvovirus B19 (B19V) infection. Historically considered the cellular receptor of B19V, the role of Gb4 and its interaction with B19V are controversial. In this study, we applied artificial viral particles, genetically modified cells, and specific competitors to address the interplay between the virus and the GSL. Our findings demonstrate that Gb4 is not involved in the binding or internalization process of the virus into permissive erythroid cells, a function that corresponds to the VP1u cognate receptor. However, Gb4 is essential at a post-internalization step before the delivery of the single-stranded viral DNA into the nucleus. In susceptible erythroid Gb4 knockout cells, incoming viruses were arrested in the endosomal compartment, showing no cytoplasmic spreading of capsids as observed in Gb4-expressing cells. Hemagglutination and binding assays revealed that pH acts as a switch to modulate the affinity between the virus and the GSL. Capsids interact with Gb4 exclusively under acidic conditions and dissociate at neutral pH. Inducing a specific Gb4-mediated attachment to permissive erythroid cells by acidification of the extracellular environment led to a non-infectious uptake of the virus, indicating that low pH-mediated binding to the GSL initiates active membrane processes resulting in vesicle formation. In summary, this study provides mechanistic insight into the interaction of B19V with Gb4. The strict pH-dependent binding to the ubiquitously expressed GSL prevents the redirection of the virus to nonpermissive tissues while promoting the interaction in acidic intracellular compartments as an essential step in infectious endocytic trafficking.


Subject(s)
Capsid/metabolism , Endocytosis/immunology , Glycosphingolipids/metabolism , Parvovirus B19, Human/genetics , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Endocytosis/physiology , Globosides/metabolism , Humans , Parvovirus B19, Human/pathogenicity , Receptors, Virus/drug effects , Receptors, Virus/metabolism , Virion/drug effects , Virion/metabolism , Virus Internalization/drug effects
5.
Asian Pac J Cancer Prev ; 22(3): 785-791, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33773542

ABSTRACT

BACKGROUND: Global trend is moving towards the use of natural phytochemicals to fight against pathogens. Human cervical cancer is directly associated with onco-potent type of Human Papilloma Virus (HPV). There is no known medicine for clearance of HPV type whose persistence is the cause of occurrence and re-occurrence of cervical cancer. The different species of fig fruit and their latex are reported to have HPV associated genital warts clearance capability. METHODS: In the current investigation, the effect of the methanol extract of Ficus benghalensis L. fruits on HPV type18 viral load in HeLa cell line was tested by doing PCR using HPV L1 primers (MY09/My011) and the cytotoxicity was also analysed by MTT assay. The induction of apoptotic activity in terms of DNA fragmentation and hyper-chromic effects of DNA was analysed. RESULTS: The PCR results showed a reduction in the HPV18 DNA and also the treatment exhibited a promising cytotoxicity with IC50 value at 211.86 µg/ml. The DNA samples from treated HeLa cells showed DNA shearing and laddering as a mark of apoptotic DNA fragmentation (Fig. 2) and the UV absorbance value at 260 nm was found to be significantly (p <0.01) higher in the DNA sample treated with fruit extract compared to the untreated DNA sample. CONCLUSION: The Ficus benghalensis L. fruit extract reduced the HPV viral load in HPV18 containing HeLa cells and showed an effective cytotoxicity on HeLa cell line. It also could induce the apoptotic activity in HeLa cell line and this study results suggest that the Ficus benghalensis L. fruits can be used to fight against cervical carcinoma, acting on HPV load.


Subject(s)
Apoptosis/drug effects , Capsid Proteins/drug effects , DNA Fragmentation/drug effects , Ficus , Human papillomavirus 18/drug effects , Phytotherapy , Plant Extracts/pharmacology , Uterine Cervical Neoplasms/drug therapy , Capsid Proteins/genetics , Cell Survival/drug effects , Female , Fruit , HeLa Cells , Human papillomavirus 18/genetics , Humans , Uterine Cervical Neoplasms/virology
6.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32907979

ABSTRACT

Pandemic human immunodeficiency virus type 1 (HIV-1) is the result of the zoonotic transmission of simian immunodeficiency virus (SIV) from the chimpanzee subspecies Pan troglodytestroglodytes (SIVcpzPtt). The related subspecies Pan troglodytesschweinfurthii is the host of a similar virus, SIVcpzPts, which did not spread to humans. We tested these viruses with small-molecule capsid inhibitors (PF57, PF74, and GS-CA1) that interact with a binding groove in the capsid that is also used by CPSF6. While HIV-1 was sensitive to capsid inhibitors in cell lines, human macrophages, and peripheral blood mononuclear cells (PBMCs), SIVcpzPtt was resistant in rhesus FRhL-2 cells and human PBMCs but was sensitive to PF74 in human HOS and HeLa cells. SIVcpzPts was insensitive to PF74 in FRhL-2 cells, HeLa cells, PBMCs, and macrophages but was inhibited by PF74 in HOS cells. A truncated version of CPSF6 (CPSF6-358) inhibited SIVcpzPtt and HIV-1, while in contrast, SIVcpzPts was resistant to CPSF6-358. Homology modeling of HIV-1, SIVcpzPtt, and SIVcpzPts capsids and binding energy estimates suggest that these three viruses bind similarly to the host proteins cyclophilin A (CYPA) and CPSF6 as well as the capsid inhibitor PF74. Cyclosporine treatment, mutation of the CYPA-binding loop in the capsid, or CYPA knockout eliminated the resistance of SIVcpzPts to PF74 in HeLa cells. These experiments revealed that the antiviral capacity of PF74 is controlled by CYPA in a virus- and cell type-specific manner. Our data indicate that SIVcpz viruses can use infection pathways that escape the antiviral activity of PF74. We further suggest that the antiviral activity of PF74 capsid inhibitors depends on cellular cofactors.IMPORTANCE HIV-1 originated from SIVcpzPtt but not from the related virus SIVcpzPts, and thus, it is important to describe molecular infection by SIVcpzPts in human cells to understand the zoonosis of SIVs. Pharmacological HIV-1 capsid inhibitors (e.g., PF74) bind a capsid groove that is also a binding site for the cellular protein CPSF6. SIVcpzPts was resistant to PF74 in HeLa cells but sensitive in HOS cells, thus indicating cell line-specific resistance. Both SIVcpz viruses showed resistance to PF74 in human PBMCs. Modulating the presence of cyclophilin A or its binding to capsid in HeLa cells overcame SIVcpzPts resistance to PF74. These results indicate that early cytoplasmic infection events of SIVcpzPts may differ between cell types and affect, in an unknown manner, the antiviral activity of capsid inhibitors. Thus, capsid inhibitors depend on the activity or interaction of currently uncharacterized cellular factors.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/chemistry , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Capsid/drug effects , Simian Immunodeficiency Virus/drug effects , mRNA Cleavage and Polyadenylation Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Binding Sites , Capsid Proteins/genetics , Cell Line , Cyclophilin A/genetics , Cyclophilin A/metabolism , Gene Knockout Techniques , HEK293 Cells , HIV-1 , HeLa Cells , Humans , Indazoles/pharmacology , Indoles/pharmacology , Leukocytes, Mononuclear/virology , Macrophages/virology , Models, Molecular , Pan troglodytes/virology , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protein Conformation , Protein Interaction Domains and Motifs , Pyridines/pharmacology , Sequence Alignment , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/chemistry , Simian Immunodeficiency Virus/genetics , Zoonoses , mRNA Cleavage and Polyadenylation Factors/genetics
7.
Proc Natl Acad Sci U S A ; 117(20): 10848-10855, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32371486

ABSTRACT

Grapevine fanleaf virus (GFLV) is a picorna-like plant virus transmitted by nematodes that affects vineyards worldwide. Nanobody (Nb)-mediated resistance against GFLV has been created recently, and shown to be highly effective in plants, including grapevine, but the underlying mechanism is unknown. Here we present the high-resolution cryo electron microscopy structure of the GFLV-Nb23 complex, which provides the basis for molecular recognition by the Nb. The structure reveals a composite binding site bridging over three domains of one capsid protein (CP) monomer. The structure provides a precise mapping of the Nb23 epitope on the GFLV capsid in which the antigen loop is accommodated through an induced-fit mechanism. Moreover, we uncover and characterize several resistance-breaking GFLV isolates with amino acids mapping within this epitope, including C-terminal extensions of the CP, which would sterically interfere with Nb binding. Escape variants with such extended CP fail to be transmitted by nematodes linking Nb-mediated resistance to vector transmission. Together, these data provide insights into the molecular mechanism of Nb23-mediated recognition of GFLV and of virus resistance loss.


Subject(s)
Nepovirus/drug effects , Plant Diseases/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Viral/immunology , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/drug effects , Cryoelectron Microscopy , Epitopes/chemistry , Models, Molecular , Nematoda/virology , Nepovirus/ultrastructure , Plant Diseases/virology , Plant Leaves/virology , Plant Viruses/immunology , Plant Viruses/physiology , Protein Conformation , Vitis
8.
Int J Antimicrob Agents ; 55(4): 105926, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32092396

ABSTRACT

Antiretroviral therapy (ART) can effectively suppress replication of human immunodeficiency virus type 1 (HIV-1) and limit disease progression. However, ART is unable to eradicate the virus, and the requirement for lifelong treatment may have side effects and may lead to the development of resistance. New approaches to prevent and treat HIV-1 infection should therefore be developed. HIV-1 capsid (CA) protein is an unexploited but attractive target for antiviral drug development. The hydrophobic cavity of the C-terminal domain of CA (CA CTD) has been validated as a potential target for antiviral drugs. Binding of compounds to this conserved non-polar groove in CA CTD allosterically disrupts the CA assembly. This study screened 2080 natural products to identify potential antiviral agents for further development to combat HIV-1 infection. From the primary screen at a fixed concentration of 50 µM, 16 compounds were found to be effective against this target. Six compounds observed in the primary screen were confirmed in dose-response experiments, and were tested against HIV-1-induced cytopathic effects. Two compounds were found to inhibit HIV-1 replication, and the most active compound - rubranol - inhibited viral replication at a moderate micromolar concentration (EC50 = 15.85 µM). The binding modes of rubranol and hirsutanonol to CA CTD were analysed by molecular docking, providing insight for the design of drugs targeting HIV-1 CA. This study reports, for the first time, identification of natural products that showed potential as anti-HIV-1 agents by targeting the conserved hydrophobic cavity of HIV-1 CA CTD.


Subject(s)
Anti-HIV Agents/pharmacology , Biological Products/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV Infections/drug therapy , HIV-1/drug effects , Virus Replication/drug effects , Capsid/drug effects , Capsid Proteins/drug effects , Cell Line , Coumaric Acids/pharmacology , Diarylheptanoids/pharmacology , Disaccharides/pharmacology , Drug Discovery/methods , Glucosides/pharmacology , Humans , Molecular Docking Simulation , Phenols/pharmacology , Virus Assembly/drug effects , Xanthones/pharmacology
9.
Antiviral Res ; 169: 104544, 2019 09.
Article in English | MEDLINE | ID: mdl-31254557

ABSTRACT

Due to its multifaceted essential roles in virus replication and extreme genetic fragility, the human immunodeficiency virus type 1 (HIV-1) capsid (CA) protein is a valued therapeutic target. However, CA is as yet unexploited clinically, as there are no antiviral agents that target it currently on the market. To facilitate the identification of potential HIV-1 CA inhibitors, we established a homogeneous time-resolved fluorescence (HTRF) assay to screen for small molecules that target a biologically active and specific binding pocket in the C-terminal domain of HIV-1 CA (CA CTD). The assay, which is based on competition of small molecules for the binding of a known CA inhibitor (CAI) to the CA CTD, exhibited a signal-to-background ratio (S/B) > 10 and a Z' value > 0.9. In a pilot screen of three kinase inhibitor libraries containing 464 compounds, we identified one compound, TX-1918, as a low micromolecular inhibitor of the HIV-1 CA CTD-CAI interaction (IC50 = 3.81 µM) that also inhibited viral replication at moderate micromolar concentration (EC50 = 15.16 µM) and inhibited CA assembly in vitro. Based on the structure of TX-1918, an additional compound with an antiviral EC50 of 6.57 µM and cellular cytotoxicity CC50 of 102.55 µM was obtained from a compound similarity search. Thus, the HTRF-based assay has properties that are suitable for screening large compound libraries to identify novel anti-HIV-1 inhibitors targeting the CA CTD.


Subject(s)
Binding, Competitive , Capsid Proteins/drug effects , Drug Evaluation, Preclinical/methods , Fluorescence , HIV-1/drug effects , High-Throughput Screening Assays/methods , Virus Assembly/drug effects , Capsid/drug effects , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Drug Liberation , Recombinant Proteins , T-Lymphocytes , Virus Replication/drug effects
10.
Nat Microbiol ; 4(6): 933-940, 2019 06.
Article in English | MEDLINE | ID: mdl-30886358

ABSTRACT

Type 1 interferon suppresses viral replication by upregulating the expression of interferon-stimulated genes with diverse antiviral properties1. The replication of human immunodeficiency virus type 1 (HIV-1) is naturally inhibited by interferon, with the steps between viral entry and chromosomal integration of viral DNA being notably susceptible2-5. The interferon-stimulated gene myxovirus resistance 2 has been defined as an effective postentry inhibitor of HIV-1, but is only partially responsible for interferon's suppressive effect6-8. Using small interfering RNA-based library screening in interferon-α-treated cells, we sought to characterize further interferon-stimulated genes that target the pre-integration phases of HIV-1 infection, and identified human tripartite-containing motif 5α (TRIM5α) as a potent anti-HIV-1 restriction factor. Human TRIM5α, in contrast with many nonhuman orthologues, has not generally been ascribed substantial HIV-1 inhibitory function, a finding attributed to ineffective recognition of cytoplasmic viral capsids by TRIM5α2,9,10. Here, we demonstrate that interferon-α-mediated stimulation of the immunoproteasome, a proteasome isoform mainly present in immune cells and distinguished from the constitutive proteasome by virtue of its different catalytic ß-subunits, as well as the proteasome activator 28 regulatory complex11-13, and the associated accelerated turnover of TRIM5α underpin the reprogramming of human TRIM5α for effective capsid-dependent inhibition of HIV-1 DNA synthesis and infection. These observations identify a mechanism for regulating human TRIM5α antiviral function in human cells and rationalize how TRIM5α participates in the immune control of HIV-1 infection.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Carrier Proteins/metabolism , HIV Infections/immunology , HIV-1/drug effects , Proteasome Endopeptidase Complex/metabolism , Antiviral Restriction Factors , Capsid/drug effects , Capsid Proteins/drug effects , Carrier Proteins/genetics , Cell Line , Gene Silencing , HEK293 Cells , Humans , Interferon-alpha/immunology , Interferon-alpha/metabolism , Myxovirus Resistance Proteins/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Virus Replication
11.
PLoS One ; 13(9): e0203069, 2018.
Article in English | MEDLINE | ID: mdl-30212479

ABSTRACT

BACKGROUND: Disulfiram (DSF), which is used to treat alcohol dependence, has been reported to have anti-cancer effects in various malignant tumors. In this study, we investigated the anti-cancer effects and mechanism of DSF in HNSCC. METHODS: Head and neck squamous carcinoma cell lines (FaDu and Hep2) were used to analyze the anti-cancer effects of DSF. The anti-cancer effects of DSF were confirmed in vivo using a xenograft tumor model. RESULTS: The anti-cancer effects of DSF in HNSCC were found to be copper (Cu) dependent. Specifically, DSF/Cu markedly inhibited HNSCC at a concentration of 1 µM. After DSF/Cu administration, production of reactive oxygen species (ROS) was remarkable starting at 0.5 µM, suggesting that the inhibitory effects of DSF/Cu on HNSCC are mediated through the formation of ROS. The levels of phospho-JNK, phospho-cJun and phospho-p38 were increased after DSF/Cu treatment while levels of phospho-Akt were decreased. These results suggested that the inhibitory effects of DSF/Cu on HNSCC cells involve ROS formation and down-regulation of Akt-signaling. Through these molecular mechanisms, DSF ultimately induce the inhibitory effects on HNSCC cell lines mainly through autophagic cell death, not apoptotic cell death. Lastly, we investigated the clinical relevance of DSF/Cu using a HNSCC xenograft animal model, which showed that tumor growth was remarkably decreased by DSF (50 mg/kg injection). CONCLUSION: In treating patients with HNSCC, DSF may contribute to improved HNSCC patient's survival. The characteristic anti-cancer effects of DSF on HNSCC may suggest new therapeutic potential for this medication in HNSCC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Disulfiram/pharmacology , Head and Neck Neoplasms/drug therapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Autophagy/physiology , Capsid Proteins/drug effects , Capsid Proteins/physiology , Cell Line, Tumor , Copper/metabolism , Drug Evaluation, Preclinical , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice, Inbred BALB C , Neoplasm Transplantation , Reactive Oxygen Species/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Burden
12.
Antiviral Res ; 158: 135-142, 2018 10.
Article in English | MEDLINE | ID: mdl-30031759

ABSTRACT

One of the most promising viral targets in current hepatitis B virus (HBV) drug development is the core protein due to its multiple roles in the viral life cycle. Here we investigated the differences in the mode of action and antiviral activity of representatives of six different capsid assembly modifier (CAM) scaffolds: three from the well-characterized scaffolds heteroarylpyrimidine (HAP), sulfamoylbenzamide (SBA), and phenylpropenamide (PPA), and three from novel scaffolds glyoxamide-pyrrolamide (GPA), pyrazolyl-thiazole (PT), and dibenzo-thiazepin-2-one (DBT). The target activity and antiviral efficacy of the different CAMs were tested in biochemical and cellular assays. Analytical size exclusion chromatography and transmission electron microscopy showed that only the HAP compound induced formation of aberrant non-capsid structures (class II mode of action), while the remaining CAMs did not affect capsid gross morphology (class I mode of action). Intracellular lysates from the HepAD38 cell line, inducibly replicating HBV, showed no reduction in the quantities of intracellular core protein or capsid after treatment with SBA, PPA, GPA, PT, or DBT compounds; however HAP-treatment led to a profound decrease in both. Additionally, immunofluorescence staining of compound-treated HepAD38 cells showed that all non-HAP CAMs led to a shift in the equilibrium of HBV core antigen (HBcAg) towards complete cytoplasmic staining, while the HAP induced accumulation of HBcAg aggregates in the nucleus. Our study demonstrates that the novel scaffolds GPA, PT, and DBT exhibit class I modes of action, alike SBA and PPA, whereas HAP remains the only scaffold belonging to class II inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Capsid/drug effects , Hepatitis B virus/drug effects , Antiviral Agents/chemistry , Benzamides/chemistry , Benzamides/pharmacology , Benzoates , Cell Line , Drug Development , Hepatitis B Core Antigens , Hepatitis B virus/metabolism , Humans , Pyrimidines/chemistry , Pyrimidines/pharmacology , Viral Core Proteins , Virus Assembly/drug effects , Virus Replication/drug effects
13.
J Virol ; 92(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29212943

ABSTRACT

Productive picornavirus infection requires the hijacking of host cell pathways to aid with the different stages of virus entry, synthesis of the viral polyprotein, and viral genome replication. Many picornaviruses, including foot-and-mouth disease virus (FMDV), assemble capsids via the multimerization of several copies of a single capsid precursor protein into a pentameric subunit which further encapsidates the RNA. Pentamer formation is preceded by co- and posttranslational modification of the capsid precursor (P1-2A) by viral and cellular enzymes and the subsequent rearrangement of P1-2A into a structure amenable to pentamer formation. We have developed a cell-free system to study FMDV pentamer assembly using recombinantly expressed FMDV capsid precursor and 3C protease. Using this assay, we have shown that two structurally different inhibitors of the cellular chaperone heat shock protein 90 (hsp90) impeded FMDV capsid precursor processing and subsequent pentamer formation. Treatment of FMDV permissive cells with the hsp90 inhibitor prior to infection reduced the endpoint titer by more than 10-fold while not affecting the activity of a subgenomic replicon, indicating that translation and replication of viral RNA were unaffected by the drug.IMPORTANCE FMDV of the Picornaviridae family is a pathogen of huge economic importance to the livestock industry due to its effect on the restriction of livestock movement and necessary control measures required following an outbreak. The study of FMDV capsid assembly, and picornavirus capsid assembly more generally, has tended to be focused upon the formation of capsids from pentameric intermediates or the immediate cotranslational modification of the capsid precursor protein. Here, we describe a system to analyze the early stages of FMDV pentameric capsid intermediate assembly and demonstrate a novel requirement for the cellular chaperone hsp90 in the formation of these pentameric intermediates. We show the added complexity involved for this process to occur, which could be the basis for a novel antiviral control mechanism for FMDV.


Subject(s)
Capsid Proteins/metabolism , Foot-and-Mouth Disease Virus/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Virus Assembly , 3C Viral Proteases , Animals , Benzoquinones/pharmacology , Capsid Proteins/drug effects , Cell Line , Cell Survival , Cell-Free System , Cricetinae , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Foot-and-Mouth Disease/metabolism , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/growth & development , HSP90 Heat-Shock Proteins/drug effects , Isoxazoles/pharmacology , Lactams, Macrocyclic/pharmacology , Protein Precursors/drug effects , Protein Precursors/metabolism , Protein Processing, Post-Translational , RNA, Viral/genetics , RNA, Viral/metabolism , Resorcinols/pharmacology , Viral Proteins/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly/genetics , Virus Assembly/physiology , Virus Replication
14.
Sci Rep ; 7(1): 17705, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29255256

ABSTRACT

Therapeutics are currently unavailable for Venezuelan equine encephalitis virus (VEEV), which elicits flu-like symptoms and encephalitis in humans, with an estimated 14% of cases resulting in neurological disease. Here we identify anti-VEEV agents using in silico structure-based-drug-design (SBDD) for the first time, characterising inhibitors that block recognition of VEEV capsid protein (C) by the host importin (IMP) α/ß1 nuclear transport proteins. From an initial screen of 1.5 million compounds, followed by in silico refinement and screening for biological activity in vitro, we identified 21 hit compounds which inhibited IMPα/ß1:C binding with IC50s as low as 5 µM. Four compounds were found to inhibit nuclear import of C in transfected cells, with one able to reduce VEEV replication at µM concentration, concomitant with reduced C nuclear accumulation in infected cells. Further, this compound was inactive against a mutant VEEV that lacks high affinity IMPα/ß1:C interaction, supporting the mode of its antiviral action to be through inhibiting C nuclear localization. This successful application of SBDD paves the way for lead optimization for VEEV antivirals, and is an exciting prospect to identify inhibitors for the many other viral pathogens of significance that require IMPα/ß1 in their infectious cycle.


Subject(s)
Capsid Proteins/drug effects , Drug Discovery/methods , Encephalitis Virus, Venezuelan Equine/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Antiviral Agents/pharmacology , Capsid , Capsid Proteins/metabolism , Cell Nucleus/metabolism , Chlorocebus aethiops , Computer Simulation , Drug Design , Encephalitis Virus, Venezuelan Equine/pathogenicity , Humans , Nucleocytoplasmic Transport Proteins/metabolism , Vero Cells , Virus Replication/drug effects , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/metabolism , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/metabolism
15.
Antiviral Res ; 146: 102-111, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28842264

ABSTRACT

Small heterocyclic molecules such as piperazine are potential pharmacotherapeutic agents and binding of these molecules to the hydrophobic pocket of capsid protein (CP) offers a new perspective for therapeutic intervention. Here, we report the crystal structure of CP from Aura virus (AVCP) in complex with piperazine at 2.2 Å resolution. Piperazine binds to the conserved hydrophobic pocket of CP where dioxane based antivirals bind. Comparative structural studies of the piperazine-bound AVCP structure with the apo, active and dioxane-bound AVCP structures provide insights into the conformational variations in the pocket. Additionally, the molecular docking studies showed that piperazine binds into the hydrophobic pocket of Chikungunya virus CP (CVCP) with more affinity than with AVCP. Furthermore, the antiviral activity of piperazine against Chikungunya virus (CHIKV) was investigated by plaque reduction and immunofluorescence assays. The AVCP-piperazine complex may serve as a lead scaffold for structure-based design of piperazine derivatives as alphaviral inhibitors. The antiviral properties of piperazine provide its usefulness for further investigations towards the development of piperazine based anti-alphaviral drugs.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Capsid/chemistry , Chikungunya virus/drug effects , Piperazines/pharmacology , Alphavirus/chemistry , Animals , Antiviral Agents/metabolism , Capsid/drug effects , Capsid Proteins/chemistry , Chlorocebus aethiops , Crystallization , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Models, Molecular , Molecular Docking Simulation , Piperazine , Piperazines/metabolism , Protein Conformation , Vero Cells
16.
Biologicals ; 43(6): 474-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26321654

ABSTRACT

This study addresses observations made in view of testing in practice the guideline in the European Pharmacopoeia (EP) on omitting the rat potency test for release of polio containing vaccines. In general, use of the guideline is valid and the D-antigen ELISA can indeed be used as an in vitro alternative for the in vivo test. However, the set-up of the ELISA is crucial and should include detection of antigenic site 1 in polio serotype 3 as destruction of that site by trypsin results in a reduced rat potency. Antigenic site 1 in polio serotype 2 may also be modified by trypsin, but the cleavage of viral protein 1 (VP1) did not affect the rat potency. Therefore, any antigenic site, except site 1, can be used for detection of polio serotype 2. It is advised to include testing of the effect of trypsin treatment in the EP-guideline. This allows polio vaccine manufacturers to check whether their in-house ELISA needs improvement.


Subject(s)
Biological Assay , Enzyme-Linked Immunosorbent Assay , Poliovirus Vaccine, Inactivated/immunology , Poliovirus/drug effects , Trypsin/pharmacology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Biosensing Techniques , Capsid Proteins/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Immunologic , Europe , Hot Temperature , Poliovirus/immunology , Poliovirus/isolation & purification , Poliovirus/pathogenicity , Practice Guidelines as Topic , Rats , Vero Cells , Virulence/drug effects
17.
J Mol Biol ; 425(18): 3378-88, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23811054

ABSTRACT

Bacteriophage lambda is one of the most exhaustively studied of the double-stranded DNA viruses. Its assembly pathway is highly conserved among the herpesviruses and many of the bacteriophages, making it an excellent model system. Despite extensive genetic and biophysical characterization of many of the lambda proteins and the assembly pathways in which they are implicated, there is a relative dearth of structural information on many of the most critical proteins involved in lambda assembly and maturation, including that of the lambda major capsid protein. Toward this end, we have utilized a combination of chemical cross-linking/mass spectrometry and computational modeling to construct a pseudo-atomic model of the lambda major capsid protein as a monomer, as well as in the context of the assembled procapsid shell. The approach described here is generalizable and can be used to provide structural models for any biological complex of interest. The procapsid structural model is in good agreement with published biochemical data indicating that procapsid expansion exposes hydrophobic surface area and that this serves to nucleate assembly of capsid decoration protein, gpD. The model further implicates additional molecular interactions that may be critical to the assembly of the capsid shell and for the stabilization of the structure by the gpD decoration protein.


Subject(s)
Bacteriophage lambda/physiology , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid/chemistry , Models, Molecular , Amino Acid Sequence , Bacteriophage lambda/chemistry , Bacteriophage lambda/drug effects , Bacteriophage lambda/ultrastructure , Capsid/drug effects , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/drug effects , Cross-Linking Reagents/pharmacology , Glycoproteins/chemistry , Glycoproteins/metabolism , Mass Spectrometry/methods , Models, Biological , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Multimerization/physiology , Protein Stability/drug effects , Protein Structure, Quaternary , Validation Studies as Topic , Virus Assembly/drug effects , Virus Assembly/physiology
18.
J Virol ; 87(5): 2835-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269800

ABSTRACT

Herpes simplex virus 2 (HSV-2) infection is still one of the common causes of sexually transmitted diseases worldwide. The prevalence of HSV strains resistant to traditional nucleoside antiviral agents has led to the development of novel antiviral drugs. Human alpha-defensin 5 (HD5), a kind of endogenous antimicrobial peptide expressed in the epithelia of the small intestine and urogenital tract, displays natural antiviral activity. Based on arginine-rich features and adaptive evolution characteristics of vertebrate defensins, we conducted a screen for HD5 derivatives with enhanced anti-HSV-2 activity by a single arginine substitution at the adaptive evolution sites. Cell protection assay and temporal antiviral studies showed that HD5 and its mutants displayed affirmatory but differential anti-HSV-2 effects in vitro by inhibiting viral adhesion and entry. Inspiringly, the E21R-HD5 mutant had significantly higher antiviral activity than natural HD5, which is possibly attributed to the stronger binding affinity of the E21R-HD5 mutant with HSV-2 capsid protein gD, indicating that E21R mutation can increase the anti-HSV-2 potency of HD5. In a mouse model of lethal HSV-2 infection, prophylactic and/or therapeutic treatment with E21R-HD5 via intravaginal instillation remarkably alleviated the symptoms and delayed disease progress and resulted in about a 1.5-fold-higher survival rate than in the HD5 group. Furthermore, the E21R variant exhibited a 2-fold-higher antiviral potency against HIV-1 over parental HD5 in vitro. This study demonstrates that arginine mutagenesis at appropriate evolution sites may significantly enhance the antiviral activity of HD5, which also paves a facile way to search for potent antiviral drugs based on natural antimicrobial peptides.


Subject(s)
Antiviral Agents/pharmacology , HIV Infections/drug therapy , Herpes Simplex/drug therapy , Herpesvirus 2, Human/drug effects , Virus Attachment/drug effects , alpha-Defensins , Amino Acid Sequence , Animals , Anti-HIV Agents/pharmacology , Capsid Proteins/drug effects , Capsid Proteins/metabolism , Chlorocebus aethiops , Evolution, Molecular , Female , HIV Infections/prevention & control , HIV-1/drug effects , Herpes Simplex/prevention & control , Herpesvirus 2, Human/physiology , Humans , Mice , Mice, Inbred BALB C , Mutagenesis , Mutation , Sequence Alignment , Vero Cells , Viral Load , alpha-Defensins/chemistry , alpha-Defensins/genetics , alpha-Defensins/metabolism , alpha-Defensins/pharmacology
19.
J Virol ; 87(5): 2587-96, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23255800

ABSTRACT

Type I interferons induce a complex transcriptional program that leads to a generalized antiviral response against a large panel of viruses, including human immunodeficiency virus type 1 (HIV-1). However, despite the fact that interferons negatively regulate HIV-1 ex vivo, a chronic interferon state is linked to the progression of AIDS and to robust viral replication, rather than protection, in vivo. To explain this apparent contradiction, we hypothesized that HIV-1 may have evolved a partial resistance to interferon, and to test this hypothesis, we analyzed the effects of alpha interferon (IFN-α) on the infectivity of HIV-1, human immunodeficiency virus type 2 (HIV-2), and rhesus monkey simian immunodeficiency virus (SIVmac). The results we obtained indicate that HIV-1 is more resistant to an IFN-α-induced response than are HIV-2 and SIVmac. Our data indicate that the accumulation of viral DNA is more compromised following the infection of IFN-α-treated cells with HIV-2 and SIVmac than with HIV-1. This defect correlates with a faster destabilization of HIV-2 viral nucleoprotein complexes (VNCs), suggesting a link between VNC destabilization and impaired viral DNA (vDNA) accumulation. The differential susceptibilities to IFN-α of the primate lentiviruses tested here do not map to the capsid protein (CA), excluding de facto a role for human tripartite motif protein isoform 5 alpha (Trim5α) in this restriction; this also suggests that an additional restriction mechanism differentially affects primate lentivirus infection. The different behaviors of HIV-1 and HIV-2 with respect to IFN-α responses may account at least in part for the differences in pathogenesis observed between these two virus types.


Subject(s)
HIV-1/physiology , HIV-2/physiology , Interferon-alpha/immunology , Simian Immunodeficiency Virus/physiology , Virus Replication , Capsid Proteins/drug effects , Cell Line, Tumor , DNA, Viral/genetics , DNA, Viral/metabolism , HEK293 Cells , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/immunology , HIV-2/immunology , HeLa Cells , Human Immunodeficiency Virus Proteins/metabolism , Humans , Macrophages/virology , Membrane Glycoproteins , Retroviridae Proteins/metabolism , Simian Immunodeficiency Virus/immunology , Viral Envelope Proteins , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
20.
J Biol Chem ; 287(29): 24473-82, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22619177

ABSTRACT

In many viruses, a precursor particle, or procapsid, is assembled and undergoes massive chemical and physical modification to produce the infectious capsid. Capsid assembly and maturation are finely tuned processes in which viral and host factors participate. We show that the precursor of the VP2 capsid protein (pVP2) of the infectious bursal disease virus (IBDV), a double-stranded RNA virus, is processed at the C-terminal domain (CTD) by a host protease, the puromycin-sensitive aminopeptidase (PurSA). The pVP2 CTD (71 residues) has an important role in determining the various conformations of VP2 (441 residues) that build the T = 13 complex capsid. pVP2 CTD activity is controlled by co- and posttranslational proteolytic modifications of different targets by the VP4 viral protease and by VP2 itself to yield the mature VP2-441 species. Puromycin-sensitive aminopeptidase is responsible for the peptidase activity that cleaves the Arg-452-Arg-453 bond to generate the intermediate pVP2-452 polypeptide. A pVP2 R453A substitution abrogates PurSA activity. We used a baculovirus-based system to express the IBDV polyprotein in insect cells and found inefficient formation of virus-like particles similar to IBDV virions, which correlates with the absence of puromycin-sensitive aminopeptidase in these cells. Virus-like particle assembly was nonetheless rescued efficiently by coexpression of chicken PurSA or pVP2-452 protein. Silencing or pharmacological inhibition of puromycin-sensitive aminopeptidase activity in cell lines permissive for IBDV replication caused a major blockade in assembly and/or maturation of infectious IBDV particles, as virus yields were reduced markedly. PurSA activity is thus essential for IBDV replication.


Subject(s)
Aminopeptidases/metabolism , Capsid Proteins/metabolism , Infectious bursal disease virus/physiology , Peptide Hydrolases/metabolism , RNA Viruses/physiology , Virus Assembly/physiology , Virus Replication/physiology , Aminopeptidases/drug effects , Animals , Capsid Proteins/drug effects , Cell Line , Dogs , Infectious bursal disease virus/drug effects , Peptide Hydrolases/drug effects , Puromycin/pharmacology , RNA Viruses/drug effects , RNA, Double-Stranded/genetics , Virus Assembly/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...