Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
BMC Genomics ; 18(1): 502, 2017 06 30.
Article in English | MEDLINE | ID: mdl-28666407

ABSTRACT

BACKGROUND: A pool of small RNA fragments (RFs) derived from diverse cellular RNAs has recently emerged as a rich source of functionally relevant molecules. Although their formation and accumulation has been connected to various stress conditions, the knowledge on RFs produced upon viral infections is very limited. Here, we applied the next generation sequencing (NGS) to characterize RFs generated in the hepatitis C virus (HCV) cell culture model (HCV-permissive Huh-7.5 cell line). RESULTS: We found that both infected and non-infected cells contained a wide spectrum of RFs derived from virtually all RNA classes. A significant fraction of identified RFs accumulated to similar levels as miRNAs. Our analysis, focused on RFs originating from constitutively expressed non-coding RNAs, revealed three major patterns of parental RNA cleavage. We found that HCV infection induced significant changes in the accumulation of low copy number RFs, while subtly altered the levels of high copy number ones. Finally, the candidate RFs potentially relevant for host-virus interactions were identified. CONCLUSIONS: Our results indicate that RFs should be considered an important component of the Huh-7.5 transcriptome and suggest that the main factors influencing the RF biogenesis are the RNA structure and RNA protection by interacting proteins. The data presented here significantly complement the existing transcriptomic, miRnomic, proteomic and metabolomic characteristics of the HCV cell culture model.


Subject(s)
Genomics , Hepacivirus/genetics , RNA, Untranslated/genetics , RNA, Viral/genetics , Cell Line , Gene Dosage/genetics , Hepacivirus/physiology , High-Throughput Nucleotide Sequencing , Humans
3.
Gut ; 66(8): 1351-1352, 2017 08.
Article in English | MEDLINE | ID: mdl-28057691
5.
Acta Biochim Pol ; 63(4): 789-798, 2016.
Article in English | MEDLINE | ID: mdl-27776194

ABSTRACT

Hepatitis C virus (HCV) infection is one of the major causes of chronic liver diseases. Unfortunately, the mechanisms of HCV infection-induced liver injury and host-virus interactions are still not well recognized. To better understand these processes we determined the changes in the host gene expression that occur during HCV infection of Huh-7.5 cells. As a result, we identified genes that may contribute to the immune and metabolic cellular responses to infection. Pathway enrichment analysis indicated that HCV induced an increased expression of genes involved in mitogen-activated protein kinases signaling, adipocytokine signaling, cell cycle and nitrogen metabolism. In addition, the enrichment analyses of processes and molecular functions revealed that the up-regulated genes were mainly implicated in the negative regulation of phosphorylation. Construction of the pathway-gene-process network enabled exploration of a much more complex landscape of molecular interactions. Consequently, several essential processes altered by HCV infection were identified: negative regulation of cell cycle, response to endoplasmic reticulum stress, response to reactive oxygen species, toll-like receptor signaling and pattern recognition receptor signaling. The analyses of genes whose expression was decreased upon HCV infection showed that the latter were engaged in the metabolism of lipids and amino acids. Moreover, we observed disturbance in the cellular antiviral defense. Altogether, our results demonstrated that HCV infection elicits host response that includes a very wide range of cellular mechanisms. Our findings significantly broaden the understanding of complex processes that accompany HCV infection. Consequently, they may be used for developing new host-oriented therapeutic strategies.


Subject(s)
Hepacivirus/physiology , Hepatitis C/metabolism , Transcriptome , Cell Line, Tumor , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Hepatitis C/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate/genetics , Metabolic Networks and Pathways , Sequence Analysis, RNA
6.
Adv Biomed Res ; 4: 13, 2015.
Article in English | MEDLINE | ID: mdl-25625119

ABSTRACT

BACKGROUND: Fusion of Hepatitis B virus surface antigen (HBsAg) to a DNA construct might be considered as a strategy to enhance cellular and cytotoxic T-lymphocytes (CTL) responses of a Hepatitis C Virus core protein (HCVcp)-based DNA vaccine comparable to that of adjuvanted protein (subunit) immunization. MATERIALS AND METHODS: pCHCORE vector harboring coding sequence of HBsAg and HCVcp (amino acids 2-120) in tandem within the pCDNA3.1 backbone was constructed. The corresponding recombinant HCVcp was also expressed and purified in Escherichia coli. Mice were immunized either by adjuvanted HCVcp (pluronic acid + protein) or by pCHCORE vector primed/protein boosted immunization regimen. The cellular immune responses (proliferation, In vivo CTL assay and IFN-γ/IL-4 ELISpot) against a strong and dominant H2-d restricted, CD8(+)-epitopic peptide (C39) (core 39-48; RRGPRLGVRA) of HCVcp were compared in immunized animals. RESULT: Proper expression of the fused protein by pCHCORE in transiently transfected HEK 293T cells and in the expected size (around 50 kDa) was confirmed by western blotting. The immunization results indicated that the pCHCORE shifted the immune responses pathway to Th1 by enhancing the IFN-γ cytokine level much higher than protein immunization while the proliferative and CTL responses were comparable (or slightly in favor of DNA immunization). CONCLUSION: Fusion of HBsAg to HCVcp in the context of a DNA vaccine modality could augment Th1-oriented cellular and CTL responses toward a protective epitope, comparable to that of HCVcp (subunit HCV vaccine) immunization.

7.
PLoS One ; 9(3): e92140, 2014.
Article in English | MEDLINE | ID: mdl-24646941

ABSTRACT

Hepatitis C virus (HCV) establishes infection using host lipid metabolism pathways that are thus considered potential targets for indirect anti-HCV strategies. HCV enters the cell via clathrin-dependent endocytosis, interacting with several receptors, and virus-cell fusion, which depends on acidic pH and the integrity of cholesterol-rich domains of the hepatocyte membrane. The ATP-binding Cassette Transporter A1 (ABCA1) mediates cholesterol efflux from hepatocytes to extracellular Apolipoprotein A1 and moves cholesterol within cell membranes. Furthermore, it generates high-density lipoprotein (HDL) particles. HDL protects against arteriosclerosis and cardiovascular disease. We show that the up-regulation of ABCA1 gene expression and its cholesterol efflux function in Huh7.5 hepatoma cells, using the liver X receptor (LXR) agonist GW3965, impairs HCV infection and decreases levels of virus produced. ABCA1-stimulation inhibited HCV cell entry, acting on virus-host cell fusion, but had no impact on virus attachment, replication, or assembly/secretion. It did not affect infectivity or properties of virus particles produced. Silencing of the ABCA1 gene and reduction of the specific cholesterol efflux function counteracted the inhibitory effect of the GW3965 on HCV infection, providing evidence for a key role of ABCA1 in this process. Impaired virus-cell entry correlated with the reorganisation of cholesterol-rich membrane microdomains (lipid rafts). The inhibitory effect could be reversed by an exogenous cholesterol supply, indicating that restriction of HCV infection was induced by changes of cholesterol content/distribution in membrane regions essential for virus-cell fusion. Stimulation of ABCA1 expression by GW3965 inhibited HCV infection of both human primary hepatocytes and isolated human liver slices. This study reveals that pharmacological stimulation of the ABCA1-dependent cholesterol efflux pathway disrupts membrane cholesterol homeostasis, leading to the inhibition of virus-cell fusion and thus HCV cell entry. Therefore besides other beneficial roles, ABCA1 might represent a potential target for HCV therapy.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Hepacivirus/physiology , Hepatitis C/genetics , Hepatitis C/virology , Up-Regulation/genetics , ATP Binding Cassette Transporter 1/metabolism , Benzoates/pharmacology , Benzylamines/pharmacology , Cell Cycle/drug effects , Cell Fusion , Cell Line, Tumor , Cholesterol/metabolism , HEK293 Cells , Hepacivirus/drug effects , Hepacivirus/pathogenicity , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver/virology , Membrane Microdomains/metabolism , RNA, Viral/metabolism , Receptors, Virus/metabolism , Up-Regulation/drug effects , Virion/drug effects , Virion/metabolism , Virus Attachment/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects
8.
PLoS One ; 6(10): e25854, 2011.
Article in English | MEDLINE | ID: mdl-22039426

ABSTRACT

Hepatitis C virus (HCV) infection is a major cause of chronic liver disease worldwide. HCV core protein is involved in nucleocapsid formation, but it also interacts with multiple cytoplasmic and nuclear molecules and plays a crucial role in the development of liver disease and hepatocarcinogenesis. The core protein is found mostly in the cytoplasm during HCV infection, but also in the nucleus in patients with hepatocarcinoma and in core-transgenic mice. HCV core contains nuclear localization signals (NLS), but no nuclear export signal (NES) has yet been identified.We show here that the aa(109-133) region directs the translocation of core from the nucleus to the cytoplasm by the CRM-1-mediated nuclear export pathway. Mutagenesis of the three hydrophobic residues (L119, I123 and L126) in the identified NES or in the sequence encoding the mature core aa(1-173) significantly enhanced the nuclear localisation of the corresponding proteins in transfected Huh7 cells. Both the NES and the adjacent hydrophobic sequence in domain II of core were required to maintain the core protein or its fragments in the cytoplasmic compartment. Electron microscopy studies of the JFH1 replication model demonstrated that core was translocated into the nucleus a few minutes after the virus entered the cell. The blockade of nucleocytoplasmic export by leptomycin B treatment early in infection led to the detection of core protein in the nucleus by confocal microscopy and coincided with a decrease in virus replication.Our data suggest that the functional NLS and NES direct HCV core protein shuttling between the cytoplasmic and nuclear compartments, with at least some core protein transported to the nucleus. These new properties of HCV core may be essential for virus multiplication and interaction with nuclear molecules, influence cell signaling and the pathogenesis of HCV infection.


Subject(s)
Hepacivirus/metabolism , Karyopherins/physiology , Nuclear Export Signals , Receptors, Cytoplasmic and Nuclear/physiology , Viral Core Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , DNA Primers , Humans , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Molecular Sequence Data , RNA Interference , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid , Viral Core Proteins/chemistry , Exportin 1 Protein
9.
PLoS One ; 6(10): e26637, 2011.
Article in English | MEDLINE | ID: mdl-22039521

ABSTRACT

A distinctive feature of HCV is that its life cycle depends on lipoprotein metabolism. Viral morphogenesis and secretion follow the very low-density lipoprotein (VLDL) biogenesis pathway and, consequently, infectious HCV in the serum is associated with triglyceride-rich lipoproteins (TRL). Lipoprotein lipase (LPL) hydrolyzes TRL within chylomicrons and VLDL but, independently of its catalytic activity, it has a bridging activity, mediating the hepatic uptake of chylomicrons and VLDL remnants. We previously showed that exogenously added LPL increases HCV binding to hepatoma cells by acting as a bridge between virus-associated lipoproteins and cell surface heparan sulfate, while simultaneously decreasing infection levels. We show here that LPL efficiently inhibits cell infection with two HCV strains produced in hepatoma cells or in primary human hepatocytes transplanted into uPA-SCID mice with fully functional human ApoB-lipoprotein profiles. Viruses produced in vitro or in vivo were separated on iodixanol gradients into low and higher density populations, and the infection of Huh 7.5 cells by both virus populations was inhibited by LPL. The effect of LPL depended on its enzymatic activity. However, the lipase inhibitor tetrahydrolipstatin restored only a minor part of HCV infectivity, suggesting an important role of the LPL bridging function in the inhibition of infection. We followed HCV cell entry by immunoelectron microscopy with anti-envelope and anti-core antibodies. These analyses demonstrated the internalization of virus particles into hepatoma cells and their presence in intracellular vesicles and associated with lipid droplets. In the presence of LPL, HCV was retained at the cell surface. We conclude that LPL efficiently inhibits HCV infection by acting on TRL associated with HCV particles through mechanisms involving its lipolytic function, but mostly its bridging function. These mechanisms lead to immobilization of the virus at the cell surface. HCV-associated lipoproteins may therefore be a promising target for the development of new therapeutic approaches.


Subject(s)
Hepacivirus/pathogenicity , Lipoprotein Lipase/metabolism , Membrane Fusion , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Lipoproteins/metabolism , Mice , Mice, SCID , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Polymerase Chain Reaction
10.
PLoS One ; 6(1): e15871, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21283512

ABSTRACT

The biological role of the protein encoded by the alternative open reading frame (core+1/ARF) of the Hepatitis C virus (HCV) genome remains elusive, as does the significance of the production of corresponding antibodies in HCV infection. We investigated the prevalence of anti-core and anti-core+1/ARFP antibodies in HCV-positive blood donors from Cambodia, using peptide and recombinant protein-based ELISAs. We detected unusual serological profiles in 3 out of 58 HCV positive plasma of genotype 1a. These patients were negative for anti-core antibodies by commercial and peptide-based assays using C-terminal fragments of core but reacted by Western Blot with full-length core protein. All three patients had high levels of anti-core+1/ARFP antibodies. Cloning of the cDNA that corresponds to the core-coding region from these sera resulted in the expression of both core and core+1/ARFP in mammalian cells. The core protein exhibited high amino-acid homology with a consensus HCV1a sequence. However, 10 identical synonymous mutations were found, and 7 were located in the aa(99-124) region of core. All mutations concerned the third base of a codon, and 5/10 represented a T>C mutation. Prediction analyses of the RNA secondary structure revealed conformational changes within the stem-loop region that contains the core+1/ARFP internal AUG initiator at position 85/87. Using the luciferase tagging approach, we showed that core+1/ARFP expression is more efficient from such a sequence than from the prototype HCV1a RNA. We provide additional evidence of the existence of core+1/ARFP in vivo and new data concerning expression of HCV core protein. We show that HCV patients who do not produce normal anti-core antibodies have unusually high levels of anti-core+1/ARFP and harbour several identical synonymous mutations in the core and core+1/ARFP coding region that result in major changes in predicted RNA structure. Such HCV variants may favour core+1/ARFP production during HCV infection.


Subject(s)
Hepacivirus/genetics , Hepatitis C Antibodies/blood , Hepatitis C/virology , Mutation , Viral Core Proteins/genetics , Hepacivirus/immunology , Hepatitis C/immunology , Humans , Nucleic Acid Conformation , Open Reading Frames , Polymorphism, Single Nucleotide , RNA, Viral/chemistry , Viral Core Proteins/immunology
11.
J Clin Virol ; 49(1): 61-4, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20621554

ABSTRACT

BACKGROUND: According to previous data, the antibodies produced during natural hepatitis C virus (HCV) infection frequently recognize amino acids 10-43 in the core protein and 1689-1740 or 1921-1940 in the non-structural 4B (NS4B) protein. The reactivity of these peptides with the corresponding antibodies has mainly been evaluated using serum samples from Western countries where HCV genotype 1 (HCV-1) is predominant, and no information is available concerning samples from sub-Saharan countries where high HCV variability has been reported. OBJECTIVE OF THIS STUDY: To evaluate the performance of HCV core and NS4B peptide-based immunoassays in the serodiagnosis of HCV infection in Cameroon subjects. STUDY DESIGN: Three core and four NS4B-based synthetic peptides derived from HCV genotypes 1b and 2a were designed and tested against a panel of 151 serum samples from Cameroon (40 positive for HCV-1, 32 for HCV-2, 39 HCV-4, and 40 HCV-negative). RESULTS: The three core peptides all demonstrated strong immunoreactivity, regardless of the HCV genotype from which they were derived, with greater than 90% and 92% sensitivity and specificity. In contrast, the NS4B-derived peptides exhibited lower sensitivities (24.3-65.8% depending on the HCV genotype) but higher specificities (100% for all four peptides tested). CONCLUSIONS: Our findings indicate that an HCV core peptide could be used for the diagnosis of chronic HCV infection. Among the NS4B peptides tested, a chimeric NS4B peptide encompassing both N- and C-terminal portions of the NS4B protein gave a much better performance than the two component N- and C-terminal peptides used individually.


Subject(s)
Hepatitis C Antibodies/blood , Hepatitis C Antigens , Hepatitis C/diagnosis , Peptide Fragments , Viral Core Proteins , Viral Nonstructural Proteins , Amino Acid Sequence , Cameroon , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/virology , Hepatitis C Antigens/chemistry , Hepatitis C Antigens/immunology , Humans , Immunoassay , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/immunology , Sensitivity and Specificity , Viral Core Proteins/chemical synthesis , Viral Core Proteins/chemistry , Viral Core Proteins/immunology , Viral Nonstructural Proteins/chemical synthesis , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/immunology
12.
PLoS One ; 5(5): e10575, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20485506

ABSTRACT

Hepatitis C virus is a poor inducer of interferon (IFN), although its structured viral RNA can bind the RNA helicase RIG-I, and activate the IFN-induction pathway. Low IFN induction has been attributed to HCV NS3/4A protease-mediated cleavage of the mitochondria-adapter MAVS. Here, we have investigated the early events of IFN induction upon HCV infection, using the cell-cultured HCV JFH1 strain and the new HCV-permissive hepatoma-derived Huh7.25.CD81 cell subclone. These cells depend on ectopic expression of the RIG-I ubiquitinating enzyme TRIM25 to induce IFN through the RIG-I/MAVS pathway. We observed induction of IFN during the first 12 hrs of HCV infection, after which a decline occurred which was more abrupt at the protein than at the RNA level, revealing a novel HCV-mediated control of IFN induction at the level of translation. The cellular protein kinase PKR is an important regulator of translation, through the phosphorylation of its substrate the eIF2alpha initiation factor. A comparison of the expression of luciferase placed under the control of an eIF2alpha-dependent (IRES(EMCV)) or independent (IRES(HCV)) RNA showed a specific HCV-mediated inhibition of eIF2alpha-dependent translation. We demonstrated that HCV infection triggers the phosphorylation of both PKR and eIF2alpha at 12 and 15 hrs post-infection. PKR silencing, as well as treatment with PKR pharmacological inhibitors, restored IFN induction in JFH1-infected cells, at least until 18 hrs post-infection, at which time a decrease in IFN expression could be attributed to NS3/4A-mediated MAVS cleavage. Importantly, both PKR silencing and PKR inhibitors led to inhibition of HCV yields in cells that express functional RIG-I/MAVS. In conclusion, here we provide the first evidence that HCV uses PKR to restrain its ability to induce IFN through the RIG-I/MAVS pathway. This opens up new possibilities to assay PKR chemical inhibitors for their potential to boost innate immunity in HCV infection.


Subject(s)
Hepacivirus/immunology , Interferons/biosynthesis , eIF-2 Kinase/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Eukaryotic Initiation Factor-2/metabolism , Hepacivirus/drug effects , Hepatitis C/immunology , Hepatitis C/virology , Humans , Kinetics , Models, Immunological , Phosphorylation/drug effects , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Substrate Specificity/drug effects , Time Factors , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , eIF-2 Kinase/antagonists & inhibitors
13.
Pol J Microbiol ; 58(2): 93-8, 2009.
Article in English | MEDLINE | ID: mdl-19824391

ABSTRACT

Hepatitis C virus (HCV) is an enveloped, single-stranded RNA virus, belonging to the Flaviviridae family. HCV infection is a major cause of chronic hepatitis worldwide, leading to steatosis, liver cirrosis and hepatocellular carcinoma. Significant advances in understanding the mechanisms of HCV infection have been made since the development of a cell culture system reproducing the complete HCV cell cycle in vitro. HCV represents a new paradigm in interactions between the virus and its target cell, the human hepatocyte, due to the central role of lipoproteins in the HCV life cycle. Very low density lipoproteins are required for virus particle assembly and secretion. Upon the release, the infectious virus circulates in the blood as triglyceride-rich particles and infects cells using lipoprotein-receptor dependent mechanisms. HCV cell entry is a multi-step process: heparan sulphate and/or low-density lipoprotein receptor are cell surface factors mediating an initial virus attachment; subsequent virus interaction with tetraspanin CD81 and the human scavenger receptor SR-BI, the main HCV receptors, triggers virus movement to the tight junctions and its uptake via Claudin-1 and occludin. Another originality of HCV is that initiation of productive infection requires dynamic microtubules. Whereas other viruses use kinesin or dynein-dependent transport, HCV exploits mechanisms driven by microtubule polymerization to efficiently infect its target cell, in which virus nucleocapsid protein might play a particular role. An improved of understanding of the cellular events involved in HCV cell entry and transport, leading to the initiation of productive HCV infection, may reveal novel targets for anti-viral interventions.


Subject(s)
Hepacivirus/physiology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Receptors, Cell Surface
14.
J Gen Virol ; 90(Pt 5): 1055-1070, 2009 May.
Article in English | MEDLINE | ID: mdl-19264629

ABSTRACT

Hepatitis C virus (HCV), a major cause of chronic liver disease, is a single-stranded positive sense virus of the family Flaviviridae. HCV cell entry is a multi-step process, involving several viral and cellular factors that trigger virus uptake into the hepatocyte. Tetraspanin CD81, human scavenger receptor SR-BI, and tight junction molecules Claudin-1 and occludin are the main receptors that mediate HCV entry. In addition, the virus may use glycosaminoglycans and/or low density receptors on host cells as initial attachment factors. A unique feature of HCV is the dependence of virus replication and assembly on host cell lipid metabolism. Most notably, during HCV assembly and release from the infected cells, virus particles associate with lipids and very-low-density lipoproteins. Thus, infectious virus circulates in patient sera in the form of triglyceride-rich particles. Consequently, lipoproteins and lipoprotein receptors play an essential role in virus uptake and the initiation of infection. This review summarizes the current knowledge about HCV receptors, mechanisms of HCV cell entry and the role of lipoproteins in this process.


Subject(s)
Hepacivirus/physiology , Hepatitis C/virology , Hepatocytes/virology , Lipoproteins/metabolism , Receptors, Virus/metabolism , Humans , Models, Biological
15.
J Biol Chem ; 284(20): 13778-13791, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19269968

ABSTRACT

Early events leading to the establishment of hepatitis C virus (HCV) infection are not completely understood. We show that intact and dynamic microtubules play a key role in the initiation of productive HCV infection. Microtubules were required for virus entry into cells, as evidenced using virus pseudotypes presenting HCV envelope proteins on their surface. Studies carried out using the recent infectious HCV model revealed that microtubules also play an essential role in early, postfusion steps of the virus cycle. Moreover, low concentrations of vinblastin and nocodazol, microtubule-affecting drugs, and paclitaxel, which stabilizes microtubules, inhibited infection, suggesting that microtubule dynamic instability and/or treadmilling mechanisms are involved in HCV internalization and early transport. By protein chip and direct core-dependent pull-down assays, followed by mass spectrometry, we identified beta- and alpha-tubulin as cellular partners of the HCV core protein. Surface plasmon resonance analyses confirmed that core directly binds to tubulin with high affinity via amino acids 2-117. The interaction of core with tubulin in vitro promoted its polymerization and enhanced the formation of microtubules. Immune electron microscopy showed that HCV core associates, at least temporarily, with microtubules polymerized in its presence. Studies by confocal microscopy showed a juxtaposition of core with microtubules in HCV-infected cells. In summary, we report that intact and dynamic microtubules are required for virus entry into cells and for early postfusion steps of infection. HCV may exploit a direct interaction of core with tubulin, enhancing microtubule polymerization, to establish efficient infection and promote virus transport and/or assembly in infected cells.


Subject(s)
Hepacivirus/physiology , Hepatitis C/metabolism , Microtubules/metabolism , Nucleocapsid Proteins/metabolism , Tubulin/metabolism , Virus Assembly/physiology , Cell Line, Tumor , Hepacivirus/ultrastructure , Hepatitis C/genetics , Hepatitis C/pathology , Humans , Microtubules/genetics , Microtubules/ultrastructure , Nucleocapsid Proteins/genetics , Tubulin/genetics
16.
Traffic ; 9(8): 1268-82, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18489704

ABSTRACT

Attachment of hepatitis C virus (HCV) core protein to lipid droplets (LDs) is linked to release of infectious progeny from infected cells. Core progressively coats the entire LD surface from a unique site on the organelle, and this process coincides with LD aggregation around the nucleus. We demonstrate that LD redistribution requires only core protein and is accompanied by reduced abundance of adipocyte differentiation-related protein (ADRP) on LD surfaces. Using small hairpin RNA technology, we show that knock down of ADRP has a similar phenotypic effect on LD redistribution. Hence, ADRP is crucial to maintain a disperse intracellular distribution of LDs. From additional experimental evidence, LDs are associated with microtubules and aggregate principally around the microtubule-organizing centre in HCV-infected cells. Disrupting the microtubule network or microinjecting anti-dynein antibody prevented core-mediated LD redistribution. Moreover, microtubule disruption reduced virus titres, implicating transport networks in virus assembly and release. We propose that the presence of core on LDs favours their movement towards the nucleus, possibly to increase the probability of interaction between sites of HCV RNA replication and virion assembly.


Subject(s)
Dyneins/chemistry , Hepacivirus/metabolism , Lipids/chemistry , Microtubules/metabolism , Viral Core Proteins/physiology , Biological Transport , Boron Compounds/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Microscopy, Fluorescence , Microtubules/chemistry , Models, Biological , Phenotype , RNA, Small Interfering/metabolism , Viral Core Proteins/chemistry , Virion/chemistry
17.
Cell Microbiol ; 9(10): 2445-56, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17517063

ABSTRACT

The host-virus interactions leading to cell infection with hepatitis C virus (HCV) are not fully understood. The tetraspanin CD-81 and human scavenger receptor SR-BI/Cla1 are major receptors mediating virus cell entry. However, HCV in patients' sera is associated with lipoproteins and infectious potential of the virus depends on lipoproteins associated to virus particles. We show here that lipoprotein lipase (LPL), targeting triglyceride-rich lipoproteins (TRL) to the liver, mediates binding and internalization of HCV to different types of cells, acting as a bridge between virus-associated lipoproteins and cell surface heparan sulfate proteoglycans (HSPG). The dimeric structure and catalytic activity of LPL are required for LPL-mediated HCV uptake to cells. Unexpectedly, exogenous LPL significantly inhibits HCVcc infection in vitro. This effect is prevented by anti-LPL antibodies and by tetrahydrolipstatin (THL) a specific inhibitor of LPL enzymatic activity. In addition, we show that antibodies directed to apolipoprotein B (ApoB)-containing lipoproteins efficiently inhibits HCVcc infection. Our findings suggest that LPL mediates HCV cell entry by a mechanism similar to hepatic clearance of TRL from the circulation, promoting a non-productive virus uptake. These data provide new insight into mechanisms of HCV cell entry and suggest that LPL could modulate HCV infectivity in vivo.


Subject(s)
Hepacivirus/physiology , Hepatitis C/virology , Lipoprotein Lipase/physiology , Animals , Apolipoproteins B/metabolism , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Dimerization , Heparan Sulfate Proteoglycans/metabolism , Humans , Lactones/pharmacology , Macrophages/metabolism , Macrophages/virology , Orlistat , Receptors, Lipoprotein/metabolism , Virus Internalization
18.
Mol Immunol ; 44(15): 3805-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17485114

ABSTRACT

Immunoglobulin GM allotypes are associated with the outcome of several infections, including hepatitis C virus (HCV) infection, but the underlying mechanisms are not known. HCV employs sophisticated strategies to evade host immunosurveillance. One such strategy might involve the scavenging of the Fc gamma domain of the anti-HCV IgG antibodies by its Fc gamma receptor-like site formed by HCV core protein, potentially interfering with the Fc gamma-mediated host defense mechanisms. We tested the hypothesis that GM allotypes modulate this viral strategy through differential binding to the core protein. Here we show that the absorbance values for binding to the HCV core protein were significantly higher for IgG1 with GM 3 allotype than that for the allelic GM 1,2,17 determinants (p=0.0003). These results provide a mechanistic explanation for the involvement of GM allotypes in the outcome of HCV infection. These findings also shed light on the possible evolutionary selective mechanism that maintains GM polymorphism.


Subject(s)
Hepacivirus/chemistry , Immunoglobulin G/metabolism , Immunoglobulin Gm Allotypes/immunology , Receptors, IgG/immunology , Viral Core Proteins/immunology , Amino Acids , Genotype , Humans , Protein Binding
19.
Biochem Biophys Res Commun ; 354(3): 641-9, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17250802

ABSTRACT

An efficient vaccine against Hepatitis C virus (HCV) infection requires induction of strong humoral and cellular responses against viral proteins. We evaluated the immunogenicity of HCV core protein (HCVcp), a prime vaccine candidate, formulated in various human compatible adjuvants. An Escherichia coli-expressed HCVcp, purified in native conditions was used for murine immunization in separate groups of: free HCVcp (Ag), Ag+C/IFA (Freunds), Ag+CpG, Ag+M720 (Montanide ISA 720), Ag+F127 (Pluronic acid) and cocktails of Ag+F127+CpG and Ag+M720+CpG. Mice immunized with M720(+CpG) developed the highest HCVcp-specific titers of total IgG, IgG1, 2a, 2b, and that of IFN-gamma and IL-4 cytokines compared to all other groups. HCVcp-specific-CTLs against relevant MHC class I peptides were detected only for Ag+M720+CpG, Ag+M720, and Ag+CpG groups and could be blocked by antimouse-CD8 antibodies. While CTLs were stable, only F127 formulated groups demonstrated detectable IgG antibodies one year post-immunization. Hence, HCVcp formulated in M720 (with a synergistic effect by inclusion of CpG) could induce balanced and strong Th1/Th2 responses with long-lived CD4(-)CD8(+) CTLs.


Subject(s)
Anti-HIV Agents/pharmacology , CpG Islands , Th1 Cells/drug effects , Th2 Cells/drug effects , Viral Core Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Base Sequence , Escherichia coli/genetics , Hepatitis C Antigens/blood , Hepatitis C Antigens/immunology , Immunoglobulin G/blood , Interferon-gamma/blood , Interleukin-4/blood , Lymphocyte Activation/immunology , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Mannitol/pharmacology , Mice , Mice, Inbred BALB C , Oleic Acids/administration & dosage , Oleic Acids/pharmacology , Th1 Cells/immunology , Th2 Cells/immunology , Viral Core Proteins/blood
20.
J Virol ; 81(8): 4244-54, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17192308

ABSTRACT

A direct involvement of the PreS domain of the hepatitis B virus (HBV) large envelope protein, and in particular amino acid residues 21 to 47, in virus attachment to hepatocytes has been suggested by many previous studies. Several PreS-interacting proteins have been identified. However, they share few common sequence motifs, and a bona fide cellular receptor for HBV remains elusive. In this study, we aimed to identify PreS-interacting motifs and to search for novel HBV-interacting proteins and the long-sought receptor. PreS fusion proteins were used as baits to screen a phage display library of random peptides. A group of PreS-binding peptides were obtained. These peptides could bind to amino acids 21 to 47 of PreS1 and shared a linear motif (W1T2X3W4W5) sufficient for binding specifically to PreS and viral particles. Several human proteins with such a motif were identified through BLAST search. Analysis of their biochemical and structural properties suggested that lipoprotein lipase (LPL), a key enzyme in lipoprotein metabolism, might interact with PreS and HBV particles. The interaction of HBV with LPL was demonstrated by in vitro binding, virus capture, and cell attachment assays. These findings suggest that LPL may play a role in the initiation of HBV infection. Identification of peptides and protein ligands corresponding to LPL that bind to the HBV envelope will offer new therapeutic strategies against HBV infection.


Subject(s)
Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/physiology , Hepatocytes/virology , Lipoprotein Lipase/metabolism , Protein Precursors/metabolism , Receptors, Virus/analysis , Amino Acid Motifs , Binding Sites , Cell Line , Humans , Peptide Library , Protein Binding , Receptors, Virus/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Virus Attachment
SELECTION OF CITATIONS
SEARCH DETAIL
...