Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Gen Virol ; 99(2): 219-229, 2018 02.
Article in English | MEDLINE | ID: mdl-29239715

ABSTRACT

Ensuring the virological safety of biologicals is challenging due to the risk of viral contamination of raw materials and cell banks, and exposure during in-process handling to known and/or emerging viral pathogens. Viruses may contaminate raw materials and biologicals intended for human or veterinary use and remain undetected until appropriate testing measures are employed. The outbreak and expansive spread of the mosquito-borne flavivirus Zika virus (ZIKV) poses challenges to screening human- and animal -derived products used in the manufacture of biologicals. Here, we report the results of an in vitro study where detector cell lines were challenged with African and Asian lineages of ZIKV. We demonstrate that this pathogen is robustly detectable by in vitro assay, thereby providing assurance of detection of ZIKV, and in turn underpinning the robustness of in vitro virology assays in safety testing of biologicals.


Subject(s)
Biological Products/standards , Drug Contamination/prevention & control , Zika Virus Infection/virology , Zika Virus/isolation & purification , Animals , Cell Line , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Humans , Risk , Vero Cells , Zika Virus Infection/transmission
2.
Biologicals ; 49: 28-32, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28751059

ABSTRACT

Emerging viruses, as potential contaminants of raw materials used in the manufacture of biologicals represent a challenge in the safety testing of biopharmaceutical products intended for human or veterinary use. Here, we report the challenge of an in vitro adventitious virus platform used in safety testing of biologicals, where a broad panel of detector cell lines was challenged to provide evidence that Schmallenberg virus is detectable by a classical reporting endpoint of cytopathic effect with Vero, BHK-21 and CHO-K1 detector cells, within typical in vitro assay timescales. We conclude that Schmallenberg virus is robustly detectable by classical in vitro viral biosafety assays.


Subject(s)
Biological Assay , Bunyaviridae Infections , Cattle Diseases , Communicable Diseases, Emerging , Orthobunyavirus , Sheep Diseases , Animals , Bunyaviridae Infections/diagnosis , Bunyaviridae Infections/veterinary , CHO Cells , Cattle , Cattle Diseases/diagnosis , Cattle Diseases/virology , Chlorocebus aethiops , Communicable Diseases, Emerging/diagnosis , Communicable Diseases, Emerging/veterinary , Communicable Diseases, Emerging/virology , Cricetinae , Cricetulus , Cytopathogenic Effect, Viral , Sheep , Sheep Diseases/diagnosis , Sheep Diseases/virology , Vero Cells
3.
Proc Natl Acad Sci U S A ; 107(46): 20057-62, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21030679

ABSTRACT

The development of HIV integrase (IN) strand transfer inhibitors (INSTIs) and our understanding of viral resistance to these molecules have been hampered by a paucity of available structural data. We recently reported cocrystal structures of the prototype foamy virus (PFV) intasome with raltegravir and elvitegravir, establishing the general INSTI binding mode. We now present an expanded set of cocrystal structures containing PFV intasomes complexed with first- and second-generation INSTIs at resolutions of up to 2.5 Å. Importantly, the improved resolution allowed us to refine the complete coordination spheres of the catalytic metal cations within the INSTI-bound intasome active site. We show that like the Q148H/G140S and N155H HIV-1 IN variants, the analogous S217H and N224H PFV INs display reduced sensitivity to raltegravir in vitro. Crystal structures of the mutant PFV intasomes in INSTI-free and -bound forms revealed that the amino acid substitutions necessitate considerable conformational rearrangements within the IN active site to accommodate an INSTI, thus explaining their adverse effects on raltegravir antiviral activity. Furthermore, our structures predict physical proximity and an interaction between HIV-1 IN mutant residues His148 and Ser/Ala140, rationalizing the coevolution of Q148H and G140S/A mutations in drug-resistant viral strains.


Subject(s)
Drug Resistance, Viral/genetics , Evolution, Molecular , HIV Integrase Inhibitors/pharmacology , Integrases/metabolism , Retroviridae/enzymology , Amino Acid Substitution/genetics , Catalytic Domain , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/metabolism , HIV-1/enzymology , HIV-1/genetics , Inhibitory Concentration 50 , Mutation/genetics , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Raltegravir Potassium
4.
Antimicrob Agents Chemother ; 56(9): 4676-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710121

ABSTRACT

Hepatitis C virus (HCV) infection is a major global health burden and is associated with an increased risk of liver cirrhosis and hepatocellular carcinoma. There remains an unmet medical need for efficacious and safe direct antivirals with complementary modes of action for combination in treatment regimens to deliver a high cure rate with a short duration of treatment for HCV patients. Here we report the in vitro inhibitory activity, mode of action, binding kinetics, and resistance profile of TMC647055, a novel and potent nonnucleoside inhibitor of the HCV NS5B RNA-dependent RNA polymerase. In vitro combination studies with an HCV NS3/4A protease inhibitor demonstrated potent suppression of HCV RNA replication, confirming the potential for combination of these two classes in the treatment of chronic HCV infection. TMC647055 is a potent nonnucleoside NS5B polymerase inhibitor of HCV replication with a promising in vitro biochemical, kinetic, and virological profile that is currently undergoing clinical evaluation.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Cell Line , Cloning, Molecular , Drug Combinations , Drug Synergism , Escherichia coli/genetics , Genes, Reporter , Hepacivirus/enzymology , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Plasmids , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transfection , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
5.
Nucleic Acids Res ; 38(6): 1902-12, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20015971

ABSTRACT

The host restriction factor Apobec3G is a cytidine deaminase that incorporates into HIV-1 virions and interferes with viral replication. The HIV-1 accessory protein Vif subverts Apobec3G by targeting it for proteasomal degradation. We propose a model in which Apobec3G N-terminal domains symmetrically interact via a head-to-head interface containing residues 122 RLYYFW 127. To validate this model and to characterize the Apobec3G-Apobec3G and the Apobec3G-Vif interactions, the mammalian protein-protein interaction trap two-hybrid technique was used. Mutations in the head-to-head interface abrogate the Apobec3G-Apobec3G interaction. All mutations that inhibit Apobec3G-Apobec3G binding also inhibit the Apobec3G-Vif interaction, indicating that the head-to head interface plays an important role in the interaction with Vif. Only the D128K, P129A and T32Q mutations specifically affect the Apobec3G-Vif association. In our model, D128, P129 and T32 cluster at the edge of the head-to-head interface, possibly forming a Vif binding site composed of two Apobec3G molecules. We propose that Vif either binds at the Apobec3G head-to-head interface or associates with an RNA-stabilized Apobec3G oligomer.


Subject(s)
Cytidine Deaminase/chemistry , vif Gene Products, Human Immunodeficiency Virus/chemistry , APOBEC-3G Deaminase , Binding Sites , Cell Line , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Cytosine Deaminase/chemistry , Dimerization , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Structural Homology, Protein , Two-Hybrid System Techniques , vif Gene Products, Human Immunodeficiency Virus/metabolism
6.
J Gen Virol ; 91(Pt 1): 122-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19793905

ABSTRACT

The cellular DEAD-box protein DDX3 was recently shown to be essential for hepatitis C virus (HCV) replication. Prior to that, we had reported that HCV core binds to DDX3 in yeast-two hybrid and transient transfection assays. Here, we confirm by co-immunoprecipitation that this interaction occurs in cells replicating the JFH1 virus. Consistent with this result, immunofluorescence staining of infected cells revealed a dramatic redistribution of cytoplasmic DDX3 by core protein to the virus assembly sites around lipid droplets. Given this close association of DDX3 with core and lipid droplets, and its involvement in virus replication, we investigated the importance of this host factor in the virus life cycle. Mutagenesis studies located a single amino acid in the N-terminal domain of JFH1 core that when changed to alanine significantly abrogated this interaction. Surprisingly, this mutation did not alter infectious virus production and RNA replication, indicating that the core-DDX3 interaction is dispensable in the HCV life cycle. Consistent with previous studies, siRNA-led knockdown of DDX3 lowered virus production and RNA replication levels of both WT JFH1 and the mutant virus unable to bind DDX3. Thus, our study shows for the first time that the requirement of DDX3 for HCV replication is unrelated to its interaction with the viral core protein.


Subject(s)
DEAD-box RNA Helicases/metabolism , Hepacivirus/physiology , Host-Pathogen Interactions , Viral Core Proteins/metabolism , Virus Replication , Amino Acid Sequence , Amino Acid Substitution/genetics , Cell Line , DEAD-box RNA Helicases/antagonists & inhibitors , Gene Knockdown Techniques , Humans , Immunoprecipitation , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Mapping , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
7.
J Virol ; 82(21): 10366-74, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18715920

ABSTRACT

Integration of viral DNA into the host chromosome is an essential step in the life cycle of retroviruses and is facilitated by the viral integrase enzyme. The first generation of integrase inhibitors recently approved or currently in late-stage clinical trials shows great promise for the treatment of human immunodeficiency virus (HIV) infection, but virus is expected to develop resistance to these drugs. Therefore, we used a novel resistance selection protocol to follow the emergence of resistant HIV in the presence of the integrase inhibitor elvitegravir (GS-9137). We find the primary resistance-conferring mutations to be Q148R, E92Q, and T66I and demonstrate that they confer a reduction in susceptibility not only to elvitegravir but also to raltegravir (MK-0518) and other integrase inhibitors. The locations of the mutations are highlighted in the catalytic sites of integrase, and we correlate the mutations with expected drug-protein contacts. In addition, mutations that do not confer reduced susceptibility when present alone (H114Y, L74M, R20K, A128T, E138K, and S230R) are also discussed in relation to their position in the catalytic core domain and their proximity to known structural features of integrase. These data broaden the understanding of antiviral resistance against integrase inhibitors and may give insight facilitating the discovery of second-generation compounds.


Subject(s)
Drug Resistance, Viral , HIV Integrase/genetics , HIV-1/drug effects , HIV-1/genetics , Integrase Inhibitors/pharmacology , Mutation, Missense , Quinolones/pharmacology , Catalytic Domain , DNA Mutational Analysis , HIV Integrase/chemistry , Humans , Models, Molecular , Molecular Structure
8.
J Biomol Screen ; 12(6): 865-74, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17644771

ABSTRACT

Fusion of host cell and human immunodeficiency virus type 1 (HIV-1) membranes is mediated by the 2 "heptad-repeat" regions of the viral gp41 protein. The collapse of the C-terminal heptad-repeat regions into the hydrophobic grooves of a coiled-coil formed by the corresponding homotrimeric N-terminal heptad-repeat regions generates a stable 6-helix bundle. This brings viral and cell membranes together for membrane fusion, facilitating viral entry. The authors developed an assay based on soluble peptides derived from the gp41 N-terminal heptad-repeat region (IQN36) as well as from the C-terminal region (C34). Both peptides were labeled with fluorophores, IQN36 with allophycocyanin (APC) and C34 with the lanthanide europium (Eu3+). Formation of the 6-helix bundle brings both fluorophores in close proximity needed for Förster resonance energy transfer (FRET). Compounds that interfere with binding of C34-Eu with IQN36-APC suppress the FRET signal. The assay was validated with various peptides and small molecules, and quenching issues were addressed. Evaluation of a diversified compound collection in a high-throughput screening campaign enabled identification of small molecules with different chemical scaffolds that inhibit this crucial intermediate in the HIV-1 entry process. This study's observations substantiate the expediency of time-resolved FRET-based assays to identify small-molecule inhibitors of protein-protein interactions.


Subject(s)
Antiviral Agents/analysis , Antiviral Agents/pharmacology , Fluorescence Resonance Energy Transfer/methods , HIV-1/drug effects , Microbial Sensitivity Tests/methods , Virus Internalization/drug effects , Amino Acid Sequence , Binding, Competitive , HIV Envelope Protein gp41/chemistry , Models, Molecular , Molecular Sequence Data , Peptide Fragments/analysis , Peptide Fragments/pharmacology , Sequence Homology, Amino Acid
9.
J Virol Methods ; 139(1): 17-23, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17034868

ABSTRACT

The use of targeting moieties is a new and exciting field of scientific research for facilitating the specific delivery of therapeutic agents in HIV-infected patients. The interaction of a potential targeting moiety with its ligand is a crucial factor in the evaluation of a targeted approach for chemotherapeutic intervention. Therefore, we have further characterized the interaction between a potential targeting agent, the monoclonal human antibody F105, and its ligand gp120, a glycoprotein expressed on the surface of HIV-1 infected cells. We demonstrate the specificity of binding and entry of F105 to infected cells. F105 was rapidly taken up into the cell and accumulated in the Golgi apparatus. Kinetic analysis of the F105-gp120 interaction revealed an equilibrium dissociation constant (K(D)) of 0.62 nM, compared with the gp120-CD4 interaction where the K(D) was determined at 35 nM. Consequently, F105 displayed a higher gp120 affinity. This was due to a slower dissociation as compared with the natural ligand. These data further underline the potential of monoclonal antibodies as targeting agents, and offer new insights into the possibility of F105 as a targeting moiety for the delivery of antiretroviral drugs to HIV-1 infected cells.


Subject(s)
Acquired Immunodeficiency Syndrome/therapy , Antibodies, Monoclonal/metabolism , HIV Envelope Protein gp120/immunology , HIV-1 , Immunoglobulin G/metabolism , Immunoglobulin kappa-Chains/metabolism , Antibodies, Monoclonal/therapeutic use , Humans
11.
Methods Mol Biol ; 1030: 25-36, 2013.
Article in English | MEDLINE | ID: mdl-23821258

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) integrase is, in addition to reverse transcriptase and protease, an important enzymatic target for antiretroviral drug development. Integrase plays a critical role in the HIV-1 life cycle coordinating the integration of the reverse-transcribed viral DNA into the host genome. This integration step is the net result of two consecutive integrase-related processes. First, integrase removes a dinucleotide from the 3' viral DNA ends in a process called 3'-processing. Next, in a process called strand transfer, the viral DNA is integrated into the host genomic DNA. Early on, biochemical assays have played a critical role in understanding the function of HIV-1 integrase and the discovery of small-molecule inhibitors. In this chapter we describe two biochemical assays to identify inhibitors of the 3'-processing and strand transfer process of HIV-1 integrase.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , HIV-1/enzymology , High-Throughput Screening Assays/methods , DNA, Viral/genetics , DNA, Viral/metabolism , HIV-1/genetics , Humans , Virus Integration/drug effects
12.
Methods Mol Biol ; 812: 295-307, 2012.
Article in English | MEDLINE | ID: mdl-22218868

ABSTRACT

The discovery of novel antivirals for HIV and HCV has been a focus of intensive research for many years. Where the inhibition of critical viral enzymes by small molecules has proven effective for many viruses, there is considerable merit in pursuing protein-protein interactions (PPIs) as targets for therapeutic intervention. The mammalian protein-protein interaction trap (MAPPIT) is a two-hybrid system used for the study of PPIs. The bait and prey proteins are linked to deficient cytokine receptor chimeras, where the bait and prey interaction and subsequent ligand stimulation restores JAK-STAT signaling, resulting in reporter gene expression controlled by a STAT3-responsive promoter. We report the use of MAPPIT as a high-throughput screening assay for the discovery of inhibitors or stimulators of the Vif-APOBEC3G interaction and the reverse transcriptase heterodimerization (RTp66-RTp51) for HIV and the NS4A-NS3 interaction for HCV.


Subject(s)
Drug Evaluation, Preclinical/methods , HIV/metabolism , Hepacivirus/metabolism , High-Throughput Screening Assays/methods , Protein Interaction Maps/drug effects , HEK293 Cells , Humans , Protein Binding/drug effects , Protein Multimerization , Protein Structure, Quaternary , Reproducibility of Results , Viral Proteins/chemistry , Viral Proteins/metabolism
13.
Antiviral Res ; 91(2): 167-76, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21669228

ABSTRACT

Raltegravir is the first integrase strand-transfer inhibitor (INSTI) approved for use in highly active antiretroviral therapy (HAART) for the management of HIV infection. Resistance to antiretrovirals can compromise the efficacy of HAART regimens. Therefore it is important to understand the emergence of resistance to RAL and cross-resistance to other INSTIs including potential second-generation INSTIs such as MK-2048. We have now studied the question of whether in vitro resistance selection (IVRS) with RAL initiated with viruses derived from clinical isolates would result in selection of resistance mutations consistent with those arising during treatment regimens with HAART containing RAL. Some correlation was observed between the primary mutations selected in vitro and during therapy, initiated with viruses with identical IN sequences. Additionally, phenotypic cross-resistance conferred by specific mutations to RAL and MK-2048 was quantified. N155H, a RAL-associated primary resistance mutation, was selected after IVRS with MK-2048, suggesting similar mechanisms of resistance to RAL and MK-2048. This was confirmed by phenotypic analysis of 766 clonal viruses harboring IN sequences isolated at the point of virological failure from 106 patients on HAART (including RAL), where mutation Q148H/K/R together with additional secondary mutations conferred reduced susceptibility to both RAL and MK-2048. A homology model of full length HIV-1 integrase complexed with viral DNA and RAL or MK-2048, based on an X-ray structure of the prototype foamy virus integrase-DNA complex, was used to explain resistance to RAL and cross-resistance to MK-2048. These findings will be important for the further discovery and profiling of next-generation INSTIs.


Subject(s)
Drug Resistance, Viral , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , Integrases/genetics , Pyrrolidinones/pharmacology , Antiretroviral Therapy, Highly Active , Cell Line , Codon/genetics , Genotype , HIV Integrase Inhibitors/chemistry , HIV-1/genetics , HIV-1/isolation & purification , HIV-1/pathogenicity , Humans , Integrases/metabolism , Microbial Sensitivity Tests/methods , Models, Molecular , Molecular Structure , Mutation , Phenotype , Plasma/virology , Pyrrolidinones/chemistry , Quinolones/chemistry , Quinolones/pharmacology , Raltegravir Potassium , Transfection
14.
PLoS One ; 6(7): e22160, 2011.
Article in English | MEDLINE | ID: mdl-21789229

ABSTRACT

BACKGROUND: Herpes simplex virus infections are highly prevalent in humans. However, the current therapeutics suffer important drawbacks such as limited results in neonates, increasing occurrence of resistance and impeded treatment of stromal infections. Remarkably, interactions of herpesviruses with human mucosa, the locus of infection, remain poorly understood and the underlying mechanisms in stromal infection remain controversial. METHODOLOGY/PRINCIPAL FINDINGS: A human model consisting of nasal respiratory mucosa explants was characterised. Viability and integrity were examined during 96 h of cultivation. HSV1-mucosa interactions were analysed. In particular, we investigated whether HSV1 is able to reach the stroma. Explant viability and integrity remained preserved. HSV1 induced rounding up and loosening of epithelial cells with very few apoptotic and necrotic cells observed. Following 16-24 h of infection, HSV1 penetrated the basement membrane and replicated in the underlying lamina propria. CONCLUSIONS/SIGNIFICANCE: This human explant model can be used to study virus-mucosa interactions and viral mucosal invasion mechanisms. Using this model, our results provide a novel insight into the HSV1 stromal invasion mechanism and for the first time directly demonstrate that HSV1 can penetrate the basement membrane.


Subject(s)
Basement Membrane/virology , Herpesvirus 1, Human/physiology , Nasal Mucosa/virology , Epithelium/virology , Fluorescence , Humans , In Situ Nick-End Labeling , Models, Biological , Nasal Mucosa/ultrastructure
15.
Virology ; 402(2): 338-46, 2010 Jul 05.
Article in English | MEDLINE | ID: mdl-20421122

ABSTRACT

Emergence of resistance to raltegravir reduces its treatment efficacy in HIV-1-infected patients. To delineate the effect of resistance mutations on viral susceptibility to integrase inhibitors, in vitro resistance selections with raltegravir and with MK-2048, an integrase inhibitor with a second-generation-like resistance profile, were performed. Mutation Q148R arose in four out of six raltegravir-selected resistant viruses. In addition, mutations Q148K and N155H were selected. In the same time frame, no mutations were selected with MK-2048. Q148H/K/R and N155H conferred resistance to raltegravir, but only minor changes in susceptibility to MK-2048. V54I, a previously unreported mutation, selected with raltegravir, was identified as a possible compensation mutation. Mechanisms by which N155H, Q148H/K/R, Y143R and E92Q confer resistance are proposed based on a structural model of integrase. These data improve the understanding of resistance against raltegravir and cross-resistance to MK-2048 and other integrase inhibitors, which will aid in the discovery of second-generation integrase inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV Integrase Inhibitors/pharmacology , HIV Integrase/genetics , HIV-1/drug effects , Mutation, Missense , Pyrrolidinones/pharmacology , Amino Acid Substitution/genetics , DNA Mutational Analysis , HIV Integrase/chemistry , HIV-1/genetics , Humans , Models, Molecular , Protein Structure, Tertiary , Raltegravir Potassium
16.
Antiviral Res ; 84(2): 142-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19699239

ABSTRACT

Selective delivery of antiretrovirals to human immunodeficiency virus (HIV) infected cells may reduce toxicities associated with long-term highly active antiretroviral therapy (HAART), may improve therapeutic compliance and delay the emergence of resistance. We developed sterically stabilized pegylated liposomes coated with targeting ligands derived from the Fab' fragment of HIV-gp120-directed monoclonal antibody F105, and evaluated these liposomes as vehicles for targeted delivery of a novel HIV-1 protease inhibitor. We demonstrated that the immunoliposomes were selectively taken up by HIV-1-infected cells and localized intracellularly, enabling the establishment of a cytoplasmic reservoir of protease inhibitor. In antiviral experiments, the drug delivered by the immunoliposomes showed greater and longer antiviral activity than comparable concentrations of free drug or drug encapsulated in non-targeted liposomes. In conclusion, by combining a targeting moiety with drug-loaded liposomes, efficient and specific uptake by non-phagocytic HIV-infected cells was facilitated, resulting in drug delivery to infected cells. This approach to targeted delivery of antiretroviral compounds may enable the design of drug regimens for patients that allow increased therapeutic adherence and less toxic treatment of HIV infection.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Envelope Protein gp120/metabolism , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Liposomes/metabolism , Liposomes/pharmacology , Virus Replication/drug effects , Cell Line , Drug Carriers/pharmacology , HIV Infections/drug therapy , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/chemistry , HIV-1/metabolism , HIV-1/physiology , Humans , Liposomes/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , T-Lymphocytes/virology
17.
PLoS One ; 2(8): e791, 2007 Aug 29.
Article in English | MEDLINE | ID: mdl-17726521

ABSTRACT

BACKGROUND: Cellular immunity plays a key role in determining the outcome of hepatitis C virus (HCV) infection, although the majority of infections become persistent. The mechanisms behind persistence are still not clear; however, the primary site of infection, the liver, may be critical. We investigated the ability of CD8+ T-cells (CTL) to recognise and kill hepatocytes under cytokine stimulation. METHODS/PRINCIPLE FINDINGS: Resting hepatocytes cell lines expressed low levels of MHC Class I, but remained susceptible to CTL cytotoxicity. IFN-alpha treatment, in vitro, markedly increased hepatocyte MHC Class I expression, however, reduced sensitivity to CTL cytotoxicity. IFN-alpha stimulated hepatocyte lines were still able to present antigen and induce IFN-gamma expression in interacting CTL. Resistance to killing was not due to the inhibition of the FASL/FAS- pathway, as stimulated hepatocytes were still susceptible to FAS-mediated apoptosis. In vitro stimulation with IFN-alpha, or the introduction of a subgenomic HCV replicon into the HepG2 line, upregulated the expression of the granzyme-B inhibitor-proteinase inhibitor 9 (PI-9). PI-9 expression was also observed in liver tissue biopsies from patients with chronic HCV infection. CONCLUSION/SIGNIFICANCE: IFN-alpha induces resistance in hepatocytes to perforin/granzyme mediate CTL killing pathways. One possible mechanism could be through the expression of the PI-9. Hindrance of CTL cytotoxicity could contribute to the chronicity of hepatic viral infections.


Subject(s)
Hepatocytes/immunology , Interferon-alpha/metabolism , T-Lymphocytes, Cytotoxic/immunology , Apoptosis , Cell Line , Cytotoxicity, Immunologic , Granzymes/metabolism , Hepatocytes/cytology , Humans
18.
Liver Int ; 25(2): 389-402, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15780065

ABSTRACT

BACKGROUND: Liver cell lines closely resembling primary hepatocyte are essential for research on hepatitis viruses and hepatocyte function. Currently used cell lines are derived from hepatic tumours and have altered gene expression. AIMS: The generation and characterisation of novel human hepatocyte lines (HHLs) derived from healthy human liver, retaining the primary hepatocyte phenotype. RESULTS: Primary hepatocytes were immortalised with Moloney's mouse leukaemia virus expressing E6 and E7 proteins of human papillomavirus, and cultures propagated long-term. All HHLs contained markers of hepatocyte and biliary phenotype (cytokeratins 7, 8, 18 and 19), Cytochrome P450 and albumin. The HHLs did not express high levels of p53 or alpha-fetoprotein. When grown in a collagen sandwich culture, or at the air-liquid interface, HHLs were maintained as monolayer whereas Huh-7 and HepG2 formed thick layers. All HHLs showed increased capacity to bind recombinant hepatitis C virus-like particles in comparison with Huh-7 and HepG2. We also demonstrate that HHLs contained active gap junctions, and that the cells respond to stimulation with IFN-alpha by upregulation of major histocompatibility complex (MHC)-I and -II. CONCLUSIONS: These HHLs retain primary hepatocyte phenotype and should be useful for investigating mechanisms of entry and replication of hepatotropic viruses, and should also be valuable in the study of hepatocyte biology and pathology.


Subject(s)
Cell Communication/immunology , Hepatocytes/cytology , Hepatocytes/immunology , Interferon-gamma/pharmacology , Binding Sites , Cell Communication/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Gap Junctions , Hepatocytes/drug effects , Humans , Male , Reference Values , Sampling Studies , Sensitivity and Specificity
19.
J Gen Virol ; 82(Pt 7): 1533-1541, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11413363

ABSTRACT

The structures of the large (L), middle (M) and small (S) versions of the envelope proteins of hepatitis B virus remain poorly characterized due to the complex nature of their conformations. Several groups have proposed transmembrane topological models depicting the lumenally and cytosolically disposed regions of these proteins. Recently, post-translational topological changes in L have been described. However, no overall differences in the topology of the S domains of the L or M, to the S protein are predicted. In this report, we investigated a previously uncharacterized anti-S monoclonal antibody (MAb), 6B1, which recognizes a conformation-sensitive epitope in S. Unlike other anti-S MAbs tested, this MAb did not recognize its epitope in the S domain of L protein. Interestingly, however, the M protein was efficiently recognized. This unique characteristic of MAb 6B1 has allowed us to study the intracellular distribution of L and S proteins. In cells expressing both L and S, L re-localized from the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) to the membrane-associated distribution of S protein indicating that L and S interact with each other. This was confirmed by immunoprecipitation assays, which also showed that the interaction between L and S results in the secretion of L protein from cells. Overall, the ability of MAb 6B1 to selectively recognize S and M, but not L, strongly points to the existence of significant topological differences in the S domain of L. The availability of this important reagent should help further our understanding of the structure of HBV surface antigens.


Subject(s)
Hepatitis B Surface Antigens/immunology , Hepatitis B virus/immunology , Viral Envelope Proteins/immunology , Antibodies, Monoclonal/immunology , Cell Line , Endoplasmic Reticulum/metabolism , Genetic Vectors , Golgi Apparatus/metabolism , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/metabolism , Microscopy, Confocal , Precipitin Tests , Protein Binding , Protein Conformation , Recombinant Proteins/immunology , Vaccinia virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
20.
J Virol ; 76(15): 7672-82, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12097581

ABSTRACT

Purification of hepatitis C virus (HCV) from sera of infected patients has proven elusive, hampering efforts to perform structure-function analysis of the viral components. Recombinant forms of the viral glycoproteins have been used instead for functional studies, but uncertainty exists as to whether they closely mimic the virion proteins. Here, we used HCV virus-like particles (VLPs) generated in insect cells infected with a recombinant baculovirus expressing viral structural proteins. Electron microscopic analysis revealed a population of pleomorphic VLPs that were at least partially enveloped with bilayer membranes and had viral glycoprotein spikes protruding from the surface. Immunogold labeling using specific monoclonal antibodies (MAbs) demonstrated these protrusions to be the E1 and E2 glycoproteins. A panel of anti-E2 MAbs was used to probe the surface topology of E2 on the VLPs and to compare the antigenicity of the VLPs with that of truncated E2 (E2(660)) or the full-length (FL) E1E2 complex expressed in mammalian cells. While most MAbs bound to all forms of antigen, a number of others showed striking differences in their abilities to recognize the various E2 forms. All MAbs directed against hypervariable region 1 (HVR-1) recognized both native and denatured E2(660) with comparable affinities, but most bound either weakly or not at all to the FL E1E2 complex or to VLPs. HVR-1 on VLPs was accessible to these MAbs only after denaturation. Importantly, a subset of MAbs specific for amino acids 464 to 475 and 524 to 535 recognized E2(660) but not VLPs or FL E1E2 complex. The antigenic differences between E2(660,) FL E1E2, and VLPs strongly point to the existence of structural differences, which may have functional relevance. Trypsin treatment of VLPs removed the N-terminal part of E2, resulting in a 42-kDa fragment. In the presence of detergent, this was further reduced to a trypsin-resistant 25-kDa fragment, which could be useful for structural studies.


Subject(s)
Antigens, Viral/immunology , Hepacivirus/metabolism , Recombination, Genetic , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Virion , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , COS Cells , Immunohistochemistry , Microscopy, Electron , Molecular Sequence Data , Spodoptera/virology , Structure-Activity Relationship , Trypsin/metabolism , Viral Envelope Proteins/genetics , Virion/immunology , Virion/metabolism , Virion/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL